Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Atherosclerosis ; 219(2): 455-62, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22051553

ABSTRACT

OBJECTIVE: Patients with familial hypercholesterolemia (FH) due mutations in the low-density lipoprotein receptor (LDLR) suffer premature aortic calcification, an effect that is age- and gene dosage-dependent and cholesterol level independent later in life. To better understand this process, we examined a murine model. METHODS: We compared chow fed Ldlr(-/-) mice to controls at 6, 12 and 18 months and on a Western diet (WD) at 6 months. Additionally, we compared controls to Ldlr(-/-) mice and transgenic mice Tg(Pcsk9) overexpressing PCSK9, which promotes LDLR degradation. Aortas were perfused-fixed, embedded in paraffin, and sections were stained with alizarin red. Micro-computerized tomography (micro-CT) was used to quantify vascular calcification. RESULTS: Ldlr(-/-) mice develop calcification in the ascending, transverse aorta and neck vessels with a distribution similar to that of human. Calcification was most prominent in 18-month-old Ldlr(-/-) mice fed a chow diet and in 6-month-old Ldlr(-/-) mice fed a WD. Interestingly, Tg(Pcsk9) mice fed a WD develop aortic calcifications as well. Histology confirmed that the calcification were predominantly sub-intimal. Marked expression of LRP5 and WNT was observed in the Ldlr(-/-) and Tg(Pcsk9) models, but not in age-matched controls. CONCLUSIONS: The two mouse models develop aortic calcification in an age- and diet-dependent manner. Abnormal regulation of the LRP5/Wnt pathway may play a role in the calcification process. Further analysis of these aortic calcification models using this micro-CT imaging technique may provide a better understanding of the link between FH and arterial calcification.


Subject(s)
Aortic Diseases/diagnostic imaging , Aortic Diseases/metabolism , Aortography/methods , Receptors, LDL/deficiency , Vascular Calcification/diagnostic imaging , Vascular Calcification/metabolism , X-Ray Microtomography , Age Factors , Aging , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Biomarkers/blood , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Disease Progression , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Paraffin Embedding , Proprotein Convertase 9 , Proprotein Convertases , Receptors, LDL/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Severity of Illness Index , Staining and Labeling , Vascular Calcification/genetics , Vascular Calcification/pathology , Wnt Proteins/metabolism
2.
Front Pharmacol ; 2: 38, 2011.
Article in English | MEDLINE | ID: mdl-21811463

ABSTRACT

Urotensin II (UII) binds to its receptor, UT, playing an important role in the heart, kidneys, pancreas, adrenal gland, and central nervous system. In the vasculature, it acts as a potent endothelium-independent vasoconstrictor and endothelium-dependent vasodilator. In disease states, however, this constriction-dilation equilibrium is disrupted. There is an upregulation of the UII system in heart disease, metabolic syndrome, and kidney failure. The increase in UII release and UT expression suggest that UII system may be implicated in the pathology and pathogenesis of these diseases by causing an increase in acyl-coenzyme A:cholesterol acyltransferase-1 (ACAT-1) activity leading to smooth muscle cell proliferation and foam cell infiltration, insulin resistance (DMII), as well as inflammation, high blood pressure, and plaque formation. Recently, UT antagonists such as SB-611812, palosuran, and most recently a piperazino-isoindolinone based antagonist have been developed in the hope of better understanding the UII system and treating its associated diseases.

3.
Peptides ; 32(5): 956-63, 2011 May.
Article in English | MEDLINE | ID: mdl-21376094

ABSTRACT

Urotensin II (UII) is a vasoactive peptide with pleiotropic activity. Interestingly, UII levels are elevated in hyperlipidemic patients, and UII induces lipase activity in some species. However, the exact role UII plays in cholesterol homeostasis remains to be elucidated. UII knockout (UII KO) mice were generated and a plasma lipoprotein profile, and hepatocytes and macrophages cholesterol uptake, storage and synthesis was determined. UII KO had a decreased LDL cholesterol profile and liver steatosis compared to wildtype mice (WT). UII KO macrophages demonstrated enhanced ACAT activity and LDL uptake in the short term (up to 4h), of which more LDL-delivered exogenously derived cholesterol was incorporated into cholesteryl ester (CE) than the WT macrophages. UII KO macrophages generated more than two times the amount of de novo endogenously synthesized cholesterol, and of this cholesterol more than two times the relative amount was esterified to CE. In comparison, results in hepatocytes demonstrated that far more exogenously derived cholesterol was incorporated into CE in the WT cells, generating almost ten times the amount of CE than UII KO. WT cells synthesize de novo almost ten times the amount of cholesterol than UIIKO, and of that cholesterol, almost two times the amount of CE in WT than UII KO hepatocytes. In addition, more ApoB lipoproteins were secreted from WT than UII KO hepatocytes. These results demonstrate a fundamental difference between macrophages and hepatocytes in terms of cholesterol homeostasis, and suggest an important role for UII in modulating cholesterol regulation.


Subject(s)
Cholesterol/metabolism , Homeostasis/drug effects , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Macrophages/metabolism , Urotensins/pharmacology , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Blotting, Western , Cells, Cultured , Cholesterol Esters/metabolism , Cholesterol, LDL/metabolism , Chromatography, High Pressure Liquid , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Mice, Knockout
4.
Peptides ; 31(8): 1511-6, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20433884

ABSTRACT

Urotensin II (UII) and urotensin-related peptide (URP) are vasoactive neuropeptides with wide ranges of action in the normal mammalian lung, including the control of smooth muscle cell proliferation. UII and URP exert their actions by binding to the G-protein coupled receptor-14 known as UT. Lymphangioleiomyomatosis (LAM) is a disease of progressive lung destruction resulting from the excessive growth of abnormal smooth muscle-like cells that exhibit markers of neural crest origin. LAM cells also exhibit inactivation of the tumor suppressor tuberin (TSC2), excessive activity of 'mammalian target of rapamycin (mTOR), and dysregulated cell growth and proliferation. In the present study we examined the expression and distribution of UII and UT in the lungs of patients with LAM. There was abundant expression of UII, URP and UT proteins in the interstitial nodular lesions of patients with LAM. By immunohistochemistry, UII, URP and UT were co-localized with HMB45, a diagnostic marker of LAM. Immunoreactivity for UII, URP and UT was also evident over the pulmonary epithelium, pulmonary vasculature and inflammatory cells. Western blotting revealed the presence of greater UT expression in the lungs of patients with LAM compared to normal human lungs. UT expression correlated with mTOR activity, as indicated by increased phosphorylation of S6 in LAM samples. These findings demonstrate for the first time the presence of UII, URP and their receptor in the lesions of patients with LAM, and suggest a possible role in the pathogenesis of the disease.


Subject(s)
Lung Neoplasms/metabolism , Lung/metabolism , Lymphangioleiomyomatosis/metabolism , Peptide Hormones/metabolism , Receptors, G-Protein-Coupled/metabolism , Up-Regulation , Urotensins/metabolism , Adult , Biomarkers, Tumor/metabolism , Blotting, Western , Humans , Immunohistochemistry , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins , Lung/blood supply , Lung/pathology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/pathology , Melanoma-Specific Antigens/metabolism , Middle Aged , Phosphorylation , Registries , Respiratory Mucosa/metabolism , Ribosomal Protein S6/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
Am J Physiol Regul Integr Comp Physiol ; 298(5): R1156-72, 2010 May.
Article in English | MEDLINE | ID: mdl-20421634

ABSTRACT

Urotensin II (UII) is an 11 amino acid cyclic peptide originally isolated from the goby fish. The amino acid sequence of UII is exceptionally conserved across most vertebrate taxa, sharing structural similarity to somatostatin. UII binds to a class of G protein-coupled receptor known as GPR14 or the urotensin receptor (UT). UII and its receptor, UT, are widely expressed throughout the cardiovascular, pulmonary, central nervous, renal, and metabolic systems. UII is generally agreed to be the most potent endogenous vasoconstrictor discovered to date. Its physiological mechanisms are similar in some ways to other potent mediators, such as endothelin-1. For example, both compounds elicit a strong vascular smooth muscle-dependent vasoconstriction via Ca(2+) release. UII also exerts a wide range of actions in other systems, such as proliferation of vascular smooth muscle cells, fibroblasts, and cancer cells. It also 1) enhances foam cell formation, chemotaxis of inflammatory cells, and inotropic and hypertrophic effects on heart muscle; 2) inhibits insulin release, modulates glomerular filtration, and release of catecholamines; and 3) may help regulate food intake and the sleep cycle. Elevated plasma levels of UII and increased levels of UII and UT expression have been demonstrated in numerous diseased conditions, including hypertension, atherosclerosis, heart failure, pulmonary hypertension, diabetes, renal failure, and the metabolic syndrome. Indeed, some of these reports suggest that UII is a marker of disease activity. As such, the UT receptor is emerging as a promising target for therapeutic intervention. Here, a concise review is given on the vast physiologic and pathologic roles of UII.


Subject(s)
Cardiovascular Diseases/physiopathology , Urotensins/physiology , Vasoconstriction/physiology , Animals , Autocrine Communication/physiology , Humans , Kidney Diseases/physiopathology , Metabolic Syndrome/physiopathology , Paracrine Communication/physiology
6.
Am J Respir Cell Mol Biol ; 42(1): 16-20, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19717811

ABSTRACT

Endothelin-1 (ET-1) plays a central role in lung fibrosis. It is released in the lung at low concentrations from the endothelium, epithelium, and vascular smooth muscle cells and orchestrates a variety of effects. In the context of wound healing, ET-1 acts with other profibrotic mediators to recruit fibroblasts and allow for their differentiation to contractile myofibroblasts. These specialized cells in turn lay down fibrotic tissue and contract at the site of lesions to restore tissue integrity. Apoptosis and reversion to quiescence ensues. However, in diseases of the lung such as idiopathic pulmonary fibrosis (IPF), the fibrotic response is uncontrolled. Progressive injury to lung tissue, isolated both temporally and geographically, is uncontrolled and eventually causes enough tissue damage to alter pulmonary architecture and compromise function. The initiating mechanisms are as of yet largely unknown; however, ET-1 has clearly emerged as a key mediator of this disease. Here, a comprehensive overview of the role of ET-1 in fibrosis is given. A guided perspective begins from the scope of its various molecular interactions to its many cellular processes, and finally to the implications of these functions in IPF.


Subject(s)
Endothelin-1/physiology , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Animals , Apoptosis , Cell Differentiation , Endothelin Receptor Antagonists , Endothelin-1/metabolism , Humans , Mice , Models, Biological , Muscle Contraction , Time Factors , Wound Healing
7.
Circ Res ; 105(7): 686-95, 19 p following 695, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19696412

ABSTRACT

RATIONALE: Expression of the vasoactive peptide Urotensin II (UII) is elevated in a number of cardiovascular diseases. OBJECTIVE: Here, we sought to determine the effect of UII receptor (UT) gene deletion in a mouse model of atherosclerosis. METHODS AND RESULTS: UT knockout (KO) mice were crossed with ApoE KO mice to generate UT/ApoE double knockout (DKO) mice. Mice were placed on a high-fat Western-type diet for 12 weeks. We evaluated the degree of atherosclerosis and hepatic steatosis by histology. In addition, serum glucose, insulin, and lipids were determined. DKO mice exhibited significantly increased atherosclerosis compared to ApoE KO mice (P<0.05). This was associated with a significant increase in serum insulin and lipids (P<0.001) but a decrease in hepatic steatosis (P<0.001). UT gene deletion led to a significant increase in systolic pressure and pulse pressure. RT-PCR and immunoblot analyses showed significant reductions in hepatic scavenger receptors, nuclear receptors, and acyl-CoA:cholesterol acyltransferase (ACAT1) expression in DKO mice. UII induced a significant increase in intracellular cholesteryl ester formation in primary mouse hepatocytes, which was blocked by the MEK inhibitor, PD98059. Hepatocytes of UTKO mice showed a significant reduction in lipoprotein uptake compared to wild-type mice. CONCLUSIONS: We propose that UT gene deletion in an ApoE-deficient background promotes downregulation of ACAT1, which in turn attenuates hepatic lipoprotein receptor-mediated uptake and lipid transporter expression. As the liver is the main organ for uptake of lipoprotein-derived lipids, DKO leads to an increase in hyperlipidemia, with a concomitant decrease in hepatic steatosis, and consequently increased atherosclerotic lesion formation. Furthermore, the hypertension associated with UT gene deletion is likely to contribute to the increased atherosclerotic burden.


Subject(s)
Aorta/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Hyperlipidemias/metabolism , Liver/metabolism , Receptors, G-Protein-Coupled/deficiency , Urotensins/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Blood Glucose/metabolism , Blood Pressure , Cells, Cultured , Cholesterol Esters/metabolism , Dietary Fats/administration & dosage , Disease Models, Animal , Fatty Liver/metabolism , Fatty Liver/prevention & control , Genotype , Hyperlipidemias/genetics , Hyperlipidemias/pathology , Hyperlipidemias/physiopathology , Insulin/blood , Lipids/blood , Liver/drug effects , Liver/pathology , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Scavenger/metabolism , Time Factors
8.
Atherosclerosis ; 204(2): 395-404, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19111831

ABSTRACT

Urotensin II (UII) and its receptor UT are upregulated in the pathological setting of various cardiovascular diseases including atherosclerosis. However, their exact role in atherosclerosis remains to be determined. In the present study we used four strains of mice; wild-type (WT), UT(+) (a transgenic strain expressing human UT driven by the alpha-smooth muscle-specific, SM22, promoter), ApoE knockout (ko), and UT(+)/ApoE ko. All animals were fed high fat diet for 12 weeks. Western blot analysis revealed a significant increase in aortic UT expression in UT(+) relative to WT mice (P<0.05). Aortas of ApoE ko mice expressed comparable UT protein level to that of UT(+). Immunohistochemistry revealed the presence of strong expression of UT and UII proteins in the atheroma of UT(+), ApoE ko and UT(+)/ApoE ko mice, particularly in foam cells. Serum cholesterol and triglyceride levels were significantly increased in ApoE ko and in UT(+)/ApoE ko but not in UT(+) mice when compared to WT mice (P<0.0001). Analysis of aortas showed a significant increase in atherosclerotic lesion in the UT(+), ApoE ko and UT(+)/ApoE ko compared to WT mice (P<0.05). Oral administration of the UT receptor antagonist SB-657510A (30 microg/Kg/day gavage) for 10 weeks in a group of ApoE ko mice fed on high fat diet resulted in a significant reduction of lesion (P<0.001). SB-657510A also significantly reduced ACAT-1 protein expression in the atherosclerotic lesion of ApoE ko mice (P<0.05). The present findings demonstrate an important role for UT in the pathogenesis of atherosclerosis. The use of UT receptor antagonists may provide a beneficial tool in the management of this debilitating disease process.


Subject(s)
Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Cardiovascular Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , Sulfonamides/pharmacology , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blotting, Western , Cholesterol, Dietary/blood , Disease Models, Animal , Foam Cells/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Promoter Regions, Genetic , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Triglycerides/blood , Urotensins/metabolism
9.
Peptides ; 29(5): 764-9, 2008 May.
Article in English | MEDLINE | ID: mdl-17988761

ABSTRACT

Urotensin-II (U-II), a cyclic undecapeptide, and its receptor, UT, have been linked to vascular and cardiac remodeling. In patients with coronary artery disease (CAD), it has been shown that U-II plasma levels are significantly greater than in normal patients and the severity of the disease is increased proportionally to the U-II plasma levels. We showed that U-II protein and mRNA levels were significantly elevated in the arteries of patients with coronary atherosclerosis in comparison to healthy arteries. We observed U-II expression in endothelial cells, foam cells, and myointimal and medial vSMCs of atherosclerotic human coronary arteries. Recent studies have demonstrated that U-II acts in synergy with mildly oxidized LDL inducing vascular smooth muscle cell (vSMC) proliferation. Additionally, U-II has been shown to induce cardiac fibrosis and cardiomyocyte hypertrophy leading to cardiac remodeling. When using a selective U-II antagonist, SB-611812, we demonstrated a decrease in cardiac dysfunction including a reduction in cardiomyocyte hypertrophy and cardiac fibrosis. These findings suggest that U-II is undoubtedly a potential therapeutic target in treating cardiovascular remodeling.


Subject(s)
Cardiovascular System , Coronary Artery Disease/metabolism , Urotensins/metabolism , Ventricular Remodeling/physiology , Cardiovascular System/anatomy & histology , Cardiovascular System/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Humans , Sulfonamides/metabolism , Urotensins/antagonists & inhibitors , Urotensins/genetics
10.
Int J Biomed Sci ; 3(1): 38-45, 2007 Mar.
Article in English | MEDLINE | ID: mdl-23675019

ABSTRACT

Treatment for symptomatic atherosclerosis is being carried out by balloon mediated angioplasty, with or without stent implantation, more and more frequently. Although advances with the development of drug eluting stents have improved prognosis, restenosis is still the most limiting factor for this treatment modality. Urotensin-II (UII), a small pleiotropic vasoactive peptide is increasingly being recognized as a contributory factor in cardiovascular diseases. We qualitatively evaluated UII immunoreactivity (IR) in three models of balloon angioplasty mediated restenosis. Specifically, we performed balloon angioplasty in the ilio-femoral arteries of New Zealand White Rabbits (NZWR) fed either a normal chow or high fat diet. In addition, UIIIR was also assessed in stent implanted abdominal aortae of NZWR fed a high fat diet. UII was constitutively expressed in the endothelium of all arterial segments evaluated. Abundant expression of UII was associated with lesion progression, particularly in myointimal cells, and less so in medial smooth muscle cells (SMC). The strongest UII-IR was observed in foam cells of animals fed a high fat diet. We demonstrate abundant expression of UII in regenerating endothelial cells and myointimal cells in vascular lesions following balloon mediated angioplasty and stent implantation in both animals fed a normal chow and high fat diet.

11.
Curr Hypertens Rep ; 8(6): 479-83, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17139807

ABSTRACT

Urotensin-II (U-II) is a vasoactive factor with pleiotropic effects. U-II exerts its activity by binding to a G-protein-coupled receptor termed UT. U-II and its receptor are highly expressed in the cardiovascular system. Increased U-II plasma levels have been reported in patients with cardiovascular disease of varying etiologies. We and others have shown that U-II and UT expression is elevated in both clinical and experimental heart failure and atherosclerosis. U-II induces cardiac fibrosis by increasing fibroblast collagen synthesis. In addition, U-II induces cardiomyocyte hypertrophy and increased vascular smooth muscle cell proliferation. We have shown that U-II antagonism using a selective U-II blocker, SB-611812 reduces neointimal thickening and increases lumen diameter in a rat restenosis model of carotid artery angioplasty. These findings suggest an important role for U-II in cardiovascular dysfunction and remodeling.


Subject(s)
Cardiovascular Diseases/physiopathology , Urotensins/physiology , Animals , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cardiovascular Diseases/blood , Cardiovascular Diseases/metabolism , Endothelium, Vascular/physiology , Humans , Hypertension/blood , Hypertension, Pulmonary/physiopathology , Immunohistochemistry , Tunica Intima/pathology , Up-Regulation/physiology , Urotensins/blood , Urotensins/metabolism
12.
Peptides ; 27(11): 2919-26, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16919371

ABSTRACT

It is now well established that urotensin-II (UII) levels are increased in several cardiovascular diseases. We previously demonstrated that UII and the UII receptor (UT) protein levels are significantly increased in the hearts of both humans and rats with congestive heart failure (CHF). We have also recently demonstrated that UII blockade, with a selective UII antagonist, improves heart function in a rat model of ischemic CHF. Here, we evaluated the attenuation of cardiac remodeling associated with UII antagonism in the same rat model of ischemic CHF. Animals were administered a specific UT receptor antagonist, SB-611812 (30 mg/kg/day, gavage), or vehicle 30 min prior to coronary artery ligation followed by daily treatment for 8 weeks. Myocardial interstitial fibrosis was analyzed by Masson's trichrome and picrosirius red staining. RT-PCR analysis was utilized for mRNA expression studies. We used Western blotting to assess levels of collagen types I and III. Mitogenic activity of UII on cultured neonatal cardiac fibroblasts was also evaluated. Following coronary ligation, SB-611812 significantly attenuated both myocardial and endocardial interstitial fibrosis, and reduced collagen type I:III ratio (P<0.01). UII induced proliferation of cardiac fibroblasts and this mitogenic effect was significantly inhibited with 1 microM of SB-611218 (P<0.05). We demonstrate here that selective blockade of UT reduces diastolic dysfunction by decreasing myocardial fibrosis post-coronary ligation in vivo, and inhibits UII-mediated fibroblast proliferation in vitro.


Subject(s)
Myocardial Ischemia/drug therapy , Myocardial Ischemia/physiopathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Sulfonamides/pharmacology , Ventricular Remodeling/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Gene Expression Profiling , Male , Myocardial Ischemia/pathology , Myocardium/pathology , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Urotensins/pharmacology
13.
J Mol Cell Cardiol ; 41(2): 285-95, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16797584

ABSTRACT

Expression of urotensin II (UII) is significantly elevated in the hearts of patients with congestive heart failure (CHF). Recent reports have also shown increased plasma levels of UII in patients with CHF, and these levels correlated with the severity of disease. We therefore hypothesized that blockade of UII signaling would improve cardiac function in a rat model of CHF. CHF was induced in rats by ligating the left coronary artery. Animals were randomized to either treatment with a specific UT receptor antagonist, SB-611812 (30 mg/kg/day, UID by gavage), or vehicle, starting either 30 min prior to coronary ligation (early treatment) or 10 days after ligation (delayed treatment). Treatment drug or vehicle was administered daily thereafter for 8 weeks. We measured cardiac function and evaluated the levels of mRNA expression for mediators of CHF. In addition, we evaluated UII and UT protein levels using immunohistochemistry and Western blotting. Cardiomyocyte hypertrophy was evaluated by measuring cardiomyocyte cross-sectional area. Animals with CHF showed increased UII and UT expression as evidenced by immunohistochemistry and Western blotting. Treatment with the SB-611812 significantly reduced overall mortality, left ventricular end-diastolic pressure by 72%, lung edema by 71%, right ventricular systolic pressure by 92%, central venous pressure by 59%, cardiomyocyte hypertrophy by 54%, and ventricular dilatation by 79% (P < 0.05). Therefore, blockade of the UT receptor reduced mortality and improved cardiac function in this model of myocardial infarction and CHF, suggesting an important role for UII in the pathogenesis of this condition.


Subject(s)
Gene Expression Regulation/drug effects , Heart Failure/prevention & control , Myocardial Infarction/prevention & control , Sulfonamides/pharmacology , Urotensins/antagonists & inhibitors , Animals , Biomarkers/metabolism , Blood Pressure , Cell Size/drug effects , Coronary Vessels , Disease Models, Animal , Heart Failure/blood , Heart Failure/complications , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Ligation , Male , Myocardial Infarction/blood , Myocardial Infarction/complications , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Inbred Lew , Urotensins/blood , Ventricular Function, Left/drug effects , Benzenesulfonamides
14.
J Immunol ; 176(8): 4950-8, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16585591

ABSTRACT

TLRs are essential mediators of host defense against infection via recognition of unique microbial structures. Recent observations indicate that TLR4, the principal receptor for bacterial LPS, may also be activated by noninfectious stimuli including host-derived molecules and environmental oxidant stress. In mice, susceptibility to ozone-induced lung permeability has been linked to the wild-type allele of TLR4, whereas deficiency of TLR4 predisposes to lethal lung injury in hyperoxia. To precisely characterize the role of lung epithelial TLR4 expression in the host response to oxidant stress, we have created an inducible transgenic mouse model that targets the human TLR4 signaling domain to the airways. Exposure of induced transgenic mice to hyperoxia revealed a significant reduction in pulmonary apoptosis compared with controls. This phenotype was associated with sustained up-regulation of antiapoptotic molecules such as heme oxygenase-1 and Bcl-2, yet only transient activation of the transcription factor NF-kappaB. Specific in vivo knockdown of pulmonary heme oxygenase-1 or Bcl-2 expression by intranasal administration of short interfering RNA blocked the effect of TLR4 signaling on hyperoxia-induced lung apoptosis. These results define a novel role for lung epithelial TLR4 as a modulator of cellular apoptosis in response to oxidant stress.


Subject(s)
Apoptosis/immunology , Hyperoxia/immunology , Hyperoxia/pathology , Lung/immunology , Lung/pathology , Toll-Like Receptor 4/metabolism , Animals , Base Sequence , Cell Line , DNA/genetics , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Hyperoxia/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Toll-Like Receptor 4/genetics
15.
Int J Cardiol ; 108(1): 31-5, 2006 Mar 22.
Article in English | MEDLINE | ID: mdl-16516696

ABSTRACT

BACKGROUND: Urotensin-II (U-II) is a vasoactive peptide with diffuse expression in human cardiomyocyte and vascular smooth muscle cells. Recent studies have reported increased plasma levels of U-II in patients with congestive heart failure. OBJECTIVE: We sought to determine the plasma levels of U-II in patients with acute coronary syndromes (ACS), stable coronary artery disease (CAD) and healthy controls. METHODS: We prospectively measured plasma U-II levels in 54 patients with ACS, 51 patients with stable coronary disease and 29 healthy volunteers. Monoclonal antibodies against U-II were generated and plasma U-II levels were determined by radioimmunoassay from extracted venous samples. RESULTS: ACS patients had significantly lower levels than patients with stable CAD and healthy controls (2.53+/-1.62 vs. 3.45+/-2.53 vs. 3.3+/-3.9 ng/ml, p=0.008, respectively). In both ACS and stable CAD patients, we found a negative relationship between plasma U-II levels and systemic arterial pressures. The correlation coefficients for systolic and mean arterial pressure were -0.272, p=0.006 and -0.209, p=0.04, respectively. CONCLUSIONS: Plasma U-II levels were significantly decreased in patients with acute coronary syndromes and related negatively to systemic arterial pressures. This finding suggests a down-regulation of U-II expression in patients with acute coronary syndromes. CONDENSED ABSTRACT: Urotensin-II (U-II) is a vasoactive peptide with diffuse staining in human cardiomyocytes and vascular smooth muscle cells. We measured plasma U-II levels in patients with acute coronary syndromes (ACS), stable coronary artery disease (CAD) and healthy controls. We observed lower U-II levels in ACS patients and a negative correlation between U-II levels and systemic arterial pressure. This finding suggests a down-regulation of U-II expression in patients with ACS.


Subject(s)
Antihypertensive Agents/blood , Coronary Artery Disease/blood , Myocardial Ischemia/blood , Urotensins/blood , Acute Disease , Aged , Biomarkers/blood , Case-Control Studies , Down-Regulation , Female , Humans , Middle Aged , Prospective Studies , Statistics, Nonparametric , Syndrome
16.
J Mol Cell Cardiol ; 39(5): 785-91, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16171813

ABSTRACT

Recent studies have postulated that the vasoactive peptide urotensin II (UII) plays a role in the control of vascular remodeling by inducing smooth muscle proliferation and fibroblast-mediated collagen deposition. The present study examined the expression of UII mRNA and immunoreactivity in rat carotid arteries before and after balloon angioplasty. In addition, the effect of UT receptor blockade was assessed in this model using a selective non-peptidic UT receptor antagonist, SB-611812. In carotid arteries of uninjured rats (naïve group), there was weak expression of UII in endothelial cells and little to no expression in vascular smooth muscle cells. At day 7, there was intimal proliferation associated with pronounced expression of UII in myointimal cells. By day 14, there was extensive intimal thickening exhibiting strong expression of UII. The contralateral arteries of all groups exhibited similar UII expression to that of naïve arteries. Animals treated with methylcellulose (vehicle) for 28 days showed a significant increase in intimal thickening compared to sham operated animals. Treatment with the SB-611812 resulted in a significant 60% reduction in intima-to-media area ratio when compared to vehicle treatment (P<0.005). These findings demonstrate upregulation of UII following balloon angioplasty, and a significant reduction in intimal lesion in response to UT receptor blockade. The present study suggests an important role for UII in the pathogenesis of restenosis following balloon angioplasty.


Subject(s)
Angioplasty, Balloon , Coronary Restenosis/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Urotensins/antagonists & inhibitors , Urotensins/metabolism , Actins/metabolism , Animals , Carotid Arteries/cytology , Humans , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
17.
Peptides ; 26(12): 2464-72, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16026900

ABSTRACT

The vasoactive peptide urotensin-II (U-II) is best known for its ability to regulate peripheral vascular and cardiac contractile function in vivo, and recent in vitro studies have suggested a role for the peptide in the control of vascular remodeling by inducing smooth muscle proliferation and fibroblast-mediated collagen deposition. Therefore, U-II may play a role in the etiology of atherosclerosis. In the present study we sought to determine the expression of U-II in coronary arteries from patients with coronary atherosclerosis and from normal control subjects, using immunohistochemistry and in situ hybridization. In normal coronary arteries, there was little expression of U-II in all types of cells. In contrast, in patients with coronary atherosclerosis, endothelial expression of U-II was significantly increased in all diseased segments (P<0.05). Greater expression of U-II was noted in endothelial cells of lesions with subendothelial inflammation or fibrofatty lesion compared with that of endothelial cells underlined by dense fibrosis or minimal intimal thickening. Myointimal cells and foam cells also expressed U-II. In most diseased segments, medial smooth muscle cells exhibited moderate expression of U-II. These findings demonstrate upregulation of U-II in endothelial, myointimal and medial smooth muscle cells of atherosclerotic human coronary arteries, and suggest a possible role for U-II in the pathogenesis of coronary atherosclerosis.


Subject(s)
Coronary Artery Disease/metabolism , Coronary Vessels/metabolism , Foam Cells/metabolism , Gene Expression Regulation , Tunica Intima/metabolism , Urotensins/biosynthesis , Adult , Coronary Artery Disease/pathology , Coronary Vessels/pathology , Female , Foam Cells/pathology , Humans , Immunohistochemistry , In Situ Hybridization , Male , Middle Aged , Tunica Intima/pathology
18.
Peptides ; 25(10): 1789-94, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15476947

ABSTRACT

Urotensin II (UII) has been found to be a potent vasoactive peptide in humans and in a number of relevant animal models of cardiovascular disease such as the mouse, rat and other non-human primates. This peptide with structural homology to somatostatin was first isolated from the urophysis of fish and was recently found to bind to an orphan receptor in mouse and human. Initially found to have potent vasoconstrictive activities in a variety of vessels from diverse species, it has also been shown to exert vasodilatation in certain vessels in the rat and human by various endothelium-dependent mechanisms. The various vasoactive properties of UII suggest that the peptide may have a physiological role in maintaining vascular tone and therefore may have a role in the pathophysiology of a number of human diseases such as heart failure. Moreover, UII has also been implicated as a mitogen of vascular smooth muscle cells suggesting a deleterious role in atherosclerosis and coronary artery disease. In addition, there is evidence to demonstrate that UII has multiple metabolic effects on cholesterol metabolism, glycemic control and hypertension and therefore may be implicated in the development of insulin resistance and the metabolic syndrome.


Subject(s)
Cardiovascular Diseases/physiopathology , Urotensins/physiology , Animals , Blood Vessels/drug effects , Blood Vessels/physiology , Humans , Insulin Resistance/physiology , Kidney Neoplasms/physiopathology , Mammals , Metabolic Syndrome/physiopathology
19.
Atherosclerosis ; 176(1): 117-23, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15306183

ABSTRACT

Urotensin II (U-II), a novel vasoactive peptide, possesses a wide range of cardiovascular effects. U-II binds a seven transmembrane spanning G-protein coupled receptor termed GPR14. In the present study, we have characterized U-II expression in both carotid and aortic atherosclerotic plaques. Using immunohistochemistry we demonstrated U-II immunoreactivity in endothelial, smooth muscle and inflammatory cells of both carotid and aortic plaques, with a clear propensity for intimal staining. Using quantitative real-time RT-PCR we observed both increased U-II and GPR14 mRNA expression in tissue extracts from abdominal aortic aneurysms. We also extended our PCR analysis to include leukocyte expression of U-II and GPR14. We found that lymphocytes were by far the largest producers of U-II mRNA. In contrast monocytes and macrophages were the largest producers of GPR14 mRNA, with relatively little expression in foam cells, lymphocytes, and platelets. Our findings qualitatively and quantitatively demonstrate increased expression of U-II in atherosclerosis with a large degree of inflammatory cell involvement. These findings suggest a possible role for U-II in the pathophysiology of atherosclerosis.


Subject(s)
Aorta/metabolism , Aortic Aneurysm, Abdominal/metabolism , Carotid Artery Diseases/metabolism , Receptors, G-Protein-Coupled/metabolism , Urotensins/metabolism , Adult , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/physiopathology , Aortic Aneurysm, Abdominal/surgery , Carotid Artery Diseases/physiopathology , Carotid Artery Diseases/surgery , Endarterectomy, Carotid , Gene Expression , Humans , Immunohistochemistry , Leukocytes/physiology , Male , Middle Aged , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Urotensins/genetics
20.
J Mol Cell Cardiol ; 37(1): 71-7, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15242737

ABSTRACT

Release of inflammatory mediators within the ischemic myocardium has long been thought to contribute to myocardial damage and dysfunction. Myocardial infarction (MI) and congestive heart failure (CHF) were induced in rats by ligating the left coronary artery. Animals were treated with the selective cyclooxygenase-2 (COX-2) inhibitor-5,5-dimethyl-3-(3-fluorophenyl1)-4-(4-methyl-sulphonyl-2(5H)-fluranone (DFU), low-, high-dose acetyl salicylic acid (aspirin), or vehicle for 3 months. Strong immunoreactivity for COX-2 was detected in the cardiomyocytes, vascular endothelial cells, and macrophages in the infarcted myocardium. Compared to the vehicle, treatment with DFU significantly reduced left ventricular end-diastolic pressure, central venous pressure, lung wet/dry ratio and infarct size, and improved cardiac contractility (P < 0.05). In comparison, treatment with low or high doses of aspirin did not significantly impact any of these parameters. These findings demonstrate that induction of myocardial COX-2 in rats with CHF secondary to MI contributes to the cardiac injury and dysfunction associated with this disease, and that therapy aimed at inhibiting this enzymatic pathway at the onset of the disease may be beneficial in the treatment of MI and CHF.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Myocardial Infarction/pathology , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Aspirin/pharmacology , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Echocardiography , Endothelium, Vascular/enzymology , Endothelium, Vascular/immunology , Furans/pharmacology , Heart Failure/enzymology , Heart Ventricles , Immunohistochemistry , Inflammation , Myocardial Infarction/enzymology , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Inbred Lew
SELECTION OF CITATIONS
SEARCH DETAIL
...