Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Arterioscler Thromb Vasc Biol ; 36(5): e41-50, 2016 05.
Article in English | MEDLINE | ID: mdl-27034473

ABSTRACT

OBJECTIVE: Thrombin signaling promotes atherosclerosis by initiating inflammatory events indirectly through platelet activation and directly via protease-activated receptors. Therefore, endogenous thrombin inhibitors may be relevant modulators of atheroprogression and cardiovascular risk. In addition, endogenous thrombin inhibitors may affect the response to non-vitamin K-dependent oral anticoagulants. Here, the question was addressed whether the small leucine-rich proteoglycan biglycan acts as an endogenous thrombin inhibitor in atherosclerosis through activation of heparin cofactor II. APPROACH AND RESULTS: Biglycan concentrations were elevated in the plasma of patients with acute coronary syndrome and in male Apolipoprotein E-deficient (ApoE(-/-)) mice. Biglycan was detected in the glycocalyx of capillaries and the subendothelial matrix of arterioles of ApoE(-/-) mice and in atherosclerotic plaques. Thereby a vascular compartment is provided that may mediate the endothelial and subendothelial activation of heparin cofactor II through biglycan. ApoE and Bgn double-deficient (ApoE(-/-)/Bgn(-/0)) mice showed higher activity of circulating thrombin, increased platelet activation and platelet adhesion in vivo, supporting a role of biglycan in balancing thrombin activity. Furthermore, concentrations of circulating cytokines and aortic macrophage content were elevated in ApoE(-/-)/Bgn(-/0) mice, suggesting a proinflammatory phenotype. Elevated platelet activation and macrophage accumulation were reversed by treating ApoE(-/-)/Bgn(-/0) mice with the thrombin inhibitor argatroban. Ultimately, ApoE(-/-)/Bgn(-/0) mice developed aggravated atherosclerosis. CONCLUSIONS: The present results indicate that biglycan plays a previously unappreciated protective role during the progression of atherosclerosis by inhibiting thrombin activity, platelet activation, and finally macrophage-mediated plaque inflammation.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Biglycan/deficiency , Inflammation/metabolism , Thrombin/metabolism , Acute Coronary Syndrome/blood , Animals , Antithrombins/pharmacology , Aorta/drug effects , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/prevention & control , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Biglycan/blood , Biglycan/genetics , Cytokines/blood , Disease Models, Animal , Genotype , Heparin Cofactor II/metabolism , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/prevention & control , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Platelet Activation , Time Factors
2.
J Mol Cell Cardiol ; 101: 145-155, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27789290

ABSTRACT

AIMS: Biglycan, a small leucine-rich proteoglycan, has been shown to play an important role in stabilizing fibrotic scars after experimental myocardial infarction. However, the role of biglycan in the development and regression of cardiomyocyte hypertrophy and fibrosis during cardiac pressure overload and unloading remains elusive. Thus, the aim of the present study was to assess the effect of biglycan on cardiac remodeling in a mouse model of left ventricular pressure overload and unloading. METHODS AND RESULTS: Left ventricular pressure overload induced by transverse aortic constriction (TAC) in mice resulted in left ventricular dysfunction, fibrosis and increased biglycan expression. Fluorescence- and magnetic-assisted sorting of cardiac cell types revealed upregulation of biglycan in the fibroblast population, but not in cardiomyocytes, endothelial cells or leukocytes after TAC. Removal of the aortic constriction (rTAC) after short-term pressure overload (3weeks) improved cardiac contractility and reversed ventricular hypertrophy but not fibrosis in wild-type (WT) mice. Biglycan ablation (KO) enhanced functional recovery but did not resolve cardiac fibrosis. After long-term TAC for 9weeks, ablation of biglycan attenuated the development of cardiac hypertrophy and fibrosis. In vitro, biglycan induced hypertrophy of neonatal rat cardiomyocytes and led to activation of a hypertrophic gene program. Putative downstream mediators of biglycan signaling include Rcan1, Abra and Tnfrsf12a. These genes were concordantly induced by TAC in WT but not in biglycan KO mice. CONCLUSIONS: Left ventricular pressure overload induces biglycan expression in cardiac fibroblasts. Ablation of biglycan improves cardiac function and attenuates left ventricular hypertrophy and fibrosis after long-term pressure overload. In vitro biglycan induces hypertrophy of cardiomyocytes, suggesting that biglycan may act as a signaling molecule between cell types to modulate cardiac remodeling.


Subject(s)
Biglycan/deficiency , Biglycan/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Ventricular Dysfunction, Left/physiopathology , Animals , Cardiomegaly/diagnosis , Disease Models, Animal , Echocardiography , Female , Fibrosis , Hypertrophy, Left Ventricular/diagnosis , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/metabolism , Male , Mice , Mice, Knockout , Myocytes, Cardiac/metabolism , Proteome , Proteomics , Rats , Ventricular Remodeling
3.
J Biol Chem ; 286(19): 17365-75, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21454527

ABSTRACT

Myocardial infarction (MI) is followed by extracellular matrix (ECM) remodeling, which is on the one hand required for the healing response and the formation of stable scar tissue. However, on the other hand, ECM remodeling can lead to fibrosis and decreased ventricular compliance. The small leucine-rich proteoglycan (SLRP), biglycan (bgn), has been shown to be critically involved in these processes. During post-infarct remodeling cardiac fibroblasts differentiate into myofibroblasts which are the main cell type mediating ECM remodeling. The aim of the present study was to characterize the role of bgn in modulating the phenotype of cardiac fibroblasts. Cardiac fibroblasts were isolated from hearts of wild-type (WT) versus bgn(-/0) mice. Phenotypic characterization of the bgn(-/0) fibroblasts revealed increased proliferation. Importantly, this phenotype of bgn(-/0) fibroblasts was abolished to the WT level by reconstitution of biglycan in the ECM. TGF-ß receptor II expression and phosphorylation of SMAD2 were increased. Furthermore, indicative of a myofibroblast phenotype bgn(-/0) fibroblasts were characterized by increased α-smooth muscle actin (α-SMA) incorporated into stress fibers, increased formation of focal adhesions, and increased contraction of collagen gels. Administration of neutralizing antibodies to TGF-ß reversed the pro-proliferative, myofibroblastic phenotype. In vivo post-MI α-SMA, TGF-ß receptor II expression, and SMAD2 phosphorylation were markedly increased in bgn(-/0) mice. Collectively, the data suggest that bgn deficiency promotes myofibroblast differentiation and proliferation in vitro and in vivo likely due to increased responses to TGF-ß and SMAD2 signaling.


Subject(s)
Biglycan/chemistry , Fibroblasts/cytology , Myocardium/cytology , Myofibroblasts/cytology , Actins/metabolism , Animals , Cell Differentiation , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Male , Mice , Mice, Transgenic , Muscle, Smooth/metabolism , Phenotype , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism
4.
BJU Int ; 110(6 Pt B): E308-17, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22500655

ABSTRACT

UNLABELLED: What's known on the subject? and What does the study add? Insulin-like growth factor mRNA-binding protein 3 (IMP3) is an oncofetal protein found to be re-expressed in a series of human cancers including bladder cancer. In vitro analyses showed an invasion and proliferation promoting effect for IMP3. Further in vitro studies suggested that IMP3 is able to bind to the mRNAs of CD44 and insulin-like growth factor 2 (IGF2), enhancing their stability and expression. However, this molecular interaction has not yet been analysed in tumour samples. In the present study, we identified for the first time high IMP3 tissue protein expression as an independent predictor of poor patients' survival in muscle-invasive bladder cancer. Furthermore, there was no correlation between IMP3 and its molecular targets in bladder carcinoma specimens and concluded that the tumour-promoting effect of IMP3 is not related to its regulatory action on IGF2 and CD44. OBJECTIVE: To assess the prognostic value and molecular actions of the oncofetal protein insulin-like growth factor mRNA-binding protein 3 (IMP3) in muscle-invasive bladder cancer (BC). PATIENTS AND METHODS: IMP3 expression was analysed by immunohistochemistry, real-time polymerase chain reaction and Western blot analysis in 224 patients with BC. The molecular targets of IMP3; CD44, insulin-like growth factor 2 (IGF2) and its receptor the IGF1 receptor (IGF1-R) were also investigated. Expression levels were correlated with clinical follow-up data by using both univariate and multivariate Cox regression analyses. RESULTS: IMP3 mRNA and protein levels were significantly elevated in high-stage and high-grade muscle-invasive BC. In muscle-invasive BC IMP3 protein but not gene expression proved to be an independent predictor of disease-specific (hazard ratio [HR] 2.58, 95% confidence interval [CI] 1.28-4.56, P = 0.004) and overall survival (HR 2.07, 95% CI 1.12-3.82, P = 0.020). The expression levels of IGF2 and CD44 showed no correlation with that of IMP3. CONCLUSIONS: High IMP3 protein levels may identify patients with BC at high risk of disease progression and may therefore select patients for a more intensive therapy or for a strict follow-up. Its high expression in high-grade bladder carcinoma cells makes IMP3 for an attractive target for therapy. The tumour promoting effect of IMP3 is independent from its regulatory action on IGF2 and CD44 expression.


Subject(s)
Insulin-Like Growth Factor Binding Protein 3/genetics , RNA, Messenger/biosynthesis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/mortality , Aged , Cells, Cultured , Female , Humans , Male , Middle Aged , Muscle, Smooth , Neoplasm Invasiveness , Survival Rate , Urinary Bladder Neoplasms/pathology
5.
J Cell Mol Med ; 15(2): 232-43, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20015203

ABSTRACT

Small leucine-rich proteoglycans (SLRPs), such as decorin and biglycan, regulate the assembly and turnover of collagenous matrix. The aim of the study was to analyse the effect of chronic rosuvastatin treatment on decorin, biglycan and the collagen matrix in ApoE-deficient mice. Twenty-week-old male ApoE-deficient mice received normal chow or 20 mg rosuvastatin/kg × day for 32 weeks. Subsequently, matrix composition was analysed by histochemistry and immunostaining at the aortic root and in innominate arteries of ApoE deficient mice as well as in human carotid endarterectomy specimens. Immunoblotting of proteoglycans was performed from aortic extracts of ApoE-deficient mice. Immunohistochemistry and immunoblotting revealed strongly increased decorin and biglycan deposition in atherosclerotic plaques at the aortic root and in innominate arteries. In contrast, versican and perlecan expression was not changed by rosuvastatin. Furthermore, matrix metalloproteinase 2 and gelatinolytic activity were decreased in response to rosuvastatin and a condensed collagen-rich matrix was formed. In carotid endarterectomy specimens of statin-treated patients increased decorin and biglycan accumulation was detected as well. Drug treatment did not change low-density lipoprotein (LDL) plasma levels in ApoE-deficient mice and did not significantly affect lipid retention at the aortic root level as demonstrated by oil-red O staining and immunohistochemistry of LDL. Long-term treatment with rosuvastatin caused pronounced remodelling of atherosclerotic plaque matrix characterized specifically by enrichment with SLRPs and formation of a condensed collagen matrix. Therefore, decorin and biglycan might represent novel targets of statin treatment that contribute to a stable plaque phenotype.


Subject(s)
Biglycan/metabolism , Decorin/metabolism , Fluorobenzenes/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Plaque, Atherosclerotic/metabolism , Proteoglycans/metabolism , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/drug therapy , Cell Line , Collagen/metabolism , Endarterectomy, Carotid , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/metabolism , Fluorobenzenes/administration & dosage , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Leucine/chemistry , Lipoproteins, LDL/analysis , Male , Mice , Mice, Knockout , Proteoglycans/chemistry , Pyrimidines/administration & dosage , Rosuvastatin Calcium , Sulfonamides/administration & dosage
6.
Circulation ; 122(22): 2313-22, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21098434

ABSTRACT

BACKGROUND: Hyaluronan is thought to mediate neointimal hyperplasia but also vasoprotection as an integral component of the endothelial glycocalyx. The present study addressed for the first time the effects of long-term pharmacological inhibition of hyaluronan synthesis on vascular function and atherosclerosis. METHODS AND RESULTS: Four-week-old apolipoprotein E-deficient mice on a Western diet received orally an inhibitor of hyaluronan synthesis, 4-methylumbelliferone (4-MU; 10 mg/g body wt), resulting in 600 nmol/L 4-MU in plasma. As a result, aortic plaque burden was markedly increased at 25 weeks. Furthermore, acetylcholine-dependent relaxation of aortic rings was decreased and mean arterial blood pressure was increased in response to 4-MU. However, hydralazine blunted the hypertensive effect of 4-MU without inhibiting the proatherosclerotic effect. A photothrombosis model revealed a prothrombotic state that was not due to increased platelet activation or increased thrombin activation as monitored by CD62P expression and the endogenous thrombin potential. Importantly, increased recruitment of macrophages to vascular lesions was detected after 2 and 21 weeks of 4-MU treatment by immunohistochemistry, by intravital microscopy, and in a peritonitis model. As a potential underlying mechanism, severe damage of the endothelial glycocalyx after 2 and 21 weeks of treatment with 4-MU was detected by electron microscopy of the innominate artery and myocardial capillaries. Furthermore, 600 nmol/L 4-MU inhibited hyaluronan synthesis in cultured endothelial cells. CONCLUSIONS: The data suggest that systemic inhibition of hyaluronan synthesis by 4-MU interferes with the protective function of the endothelial glycocalyx, thereby facilitating leukocyte adhesion, subsequent inflammation, and progression of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Disease Progression , Hyaluronic Acid/antagonists & inhibitors , Hyaluronic Acid/metabolism , Acetylcholine/pharmacology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/physiopathology , Blood Pressure/drug effects , Blood Pressure/physiology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Female , Glycocalyx/drug effects , Glycocalyx/metabolism , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Mice , Mice, Knockout , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology
7.
Basic Res Cardiol ; 105(1): 29-38, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19701787

ABSTRACT

Accumulation of biglycan, a small leucine-rich proteoglycan, in the neointima precedes the retention of lipids and accumulation of macrophages during early atherosclerosis. Biglycan is therefore considered a pro-atherogenic proteoglycan that might play a key role in atherogenesis. On the other hand biglycan ensures in part establishment of stable collagen networks. Aim of the present study was to determine whether telmisartan affects biglycan accumulation in a murine model of accelerated atherosclerosis and whether collagen matrix is affected. ApoE(-/-)-mice on Western diet were chronically (12 weeks) treated either with telmisartan (10 mg/kg) or hydralazine (500 mg/l drinking water) and systolic arterial blood pressure was determined by tail cuff plethysmography. Animals were killed and aortic plaque score, plaque morphology and extracellular matrix as well as cellular plaque composition were analyzed at the aortic root. Furthermore, expression of biglycan and enzymes involved in collagen cross-linking were analyzed in the aorta. Telmisartan and hydralazine lowered systolic arterial blood pressure to the same extent. Biglycan accumulation in the aorta and the aortic root was significantly reduced by telmisartan but not by hydralazine. The amount of collagen and collagen fibril density, macrophages and SMCs was not affected by either treatment as determined by analysis of picrosirius red staining, mac2 and alpha-SM-actin. Furthermore, telmisartan inhibited aortic plaque score and aortic root plaque size compared to mice receiving hydralazine and untreated controls. The current study shows that telmisartan reduces biglycan accumulation and inhibits atherosclerosis independently of blood pressure lowering and without affecting the collagenous plaque matrix. Thus, biglycan is a pleiotropic target of telmisartan that might contribute to the anti-atherogenic effects of this AT1-antagonist.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Atherosclerosis/drug therapy , Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Extracellular Matrix Proteins/metabolism , Proteoglycans/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Benzimidazoles/pharmacology , Benzoates/pharmacology , Biglycan , Blood Pressure/drug effects , Collagen/metabolism , Lipid Metabolism , Male , Mice , Mice, Knockout , Telmisartan
8.
Basic Res Cardiol ; 105(1): 99-108, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19701788

ABSTRACT

Biglycan, a small leucine-rich proteoglycan, is essential for scar formation and preservation of hemodynamic function after myocardial infarction, as shown in biglycan-knockout mice. Because of this important role in cardiac pathophysiology, we aimed to identify regulators of biglycan expression and posttranslational modifications in cardiac fibroblasts. Cardiac fibroblasts were isolated from neonatal Wistar-Kyoto rats and used in the first passage. Expression of biglycan was analyzed after metabolic labeling with [(35)S]-sulfate by SDS-polyacrylamide gel electrophoresis and molecular sieve chromatography; mRNA expression was examined by Northern analysis and real-time RT-PCR. Serum, thrombin, transforming growth factor beta1 (TGFbeta 1) and platelet-derived growth factor BB (PDGF-BB) strongly increased [(35)S]-labeled proteoglycan levels. Tumor necrosis factor alpha further increased the stimulatory effect of PDGF-BB. PDGF-BB increased glycosaminoglycan (GAG) chain length as shown by molecular sieve chromatography after beta-elimination to release GAG chains. Nitric oxide was the only negative regulator of biglycan as evidenced by marked downregulation in response to DETA-NO ((Z)-1-[2-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate), a long acting nitric oxide donor and SNAP (S-nitroso-N-acetyl-l,l-penicillamine), which completely inhibited PDGF-BB-induced secretion of total [(35)S]-labeled proteoglycans and biglycan mRNA expression. Of note, the molecular weight of biglycan GAG chains was even further increased by NO donors compared to control and PDGF-BB stimulation. The current results suggest that in cardiac fibroblasts, biglycan is induced by a variety of stimuli including serum, thrombin and growth factors such as PDGF-BB and TGFbeta1. This response is counteracted by NO and enhanced by TNFalpha. Interestingly, both up- and downregulation were associated with posttranslational increase of GAG chain length.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Protein Processing, Post-Translational , Proteoglycans/metabolism , Animals , Animals, Newborn , Becaplermin , Biglycan , Cells, Cultured , Glycosaminoglycans/metabolism , Nitric Oxide/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Rats , Rats, Inbred WKY , Thrombin/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
PLoS One ; 8(9): e75681, 2013.
Article in English | MEDLINE | ID: mdl-24069434

ABSTRACT

UNLABELLED: Hyaluronan (HA) is a carbohydrate of the extracellular matrix with tumor promoting effects in a variety of cancers. The present study addressed the role of HA matrix for progression and prognosis of human bladder cancer by studying the expression and function of HA-related genes. METHODS: Tissue samples of 120 patients with different stages of transitional cell bladder cancer, who underwent surgical treatment for bladder cancer at the University Hospital of Essen were analysed. mRNA-expression levels of HA synthases (HAS1-3) and HA-receptors (RHAMM and CD44) were evaluated by real time RT-PCR in comparison to healthy bladder tissue as control. In uni- and multivariate cox proportional hazard survival regression analysis, the impact of the gene expression levels on survival was assessed. In vitro knock-down of RHAMM, CD44 and HAS isoenzymes was achieved by siRNA and lentiviral shRNA in J82 bladder cancer cells. Transfected cells were analysed in vitro with regard to proliferation, cell cycle and apoptosis. J82 cells after knock-down of RHAMM were xenografted into male nu/nu athymic mice to monitor tumor progression in vivo. RESULTS: In invasive tumor stages RHAMM-, HAS1 and HAS2 mRNA-expression levels were elevated whereas HAS3v1 was reduced as compared to non-invasive tumors. Subsequently, Kaplan-Meier analysis revealed reduced bladder cancer specific survival in patients with high RHAMM mRNA and low HAS3v1 expression. Elevated RHAMM in invasive tumors was confirmed by RHAMM immunohistochemistry. Furthermore, multivariate analysis revealed that only RHAMM expression was associated with poor prognosis independent from other survival factors (HR=2.389, 95% CI 1.227-4.651, p=0.01). Lentiviral RHAMM knock-down revealed reduced J82 cell proliferation in vitro and reduced xenograft tumor growth in vivo. CONCLUSION: The data suggest that RHAMM plays a crucial role in mediating progression of muscle-invasive bladder cancer and recommends RHAMM for further evaluation as a prognostic marker or therapeutic target in bladder cancer therapy.


Subject(s)
Carcinoma, Transitional Cell/genetics , Extracellular Matrix Proteins/genetics , Hyaluronan Receptors/genetics , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , Animals , Apoptosis , Carcinoma, Transitional Cell/metabolism , Carcinoma, Transitional Cell/mortality , Carcinoma, Transitional Cell/pathology , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/metabolism , Female , Gene Knockdown Techniques , Heterografts , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Immunohistochemistry , Immunophenotyping , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Staging , Phenotype , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors , Tumor Burden/genetics , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL