Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Europace ; 23(5): 682-690, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33319222

ABSTRACT

AIMS: Adiposity plays a key role in the pathogenesis of atrial fibrillation (AF). Our aim was to study the sex differences in adipokines levels according to AF burden. METHODS AND RESULTS: Two independent cohorts of patients were studied: (i) consecutive patients with AF undergoing catheter ablation (n = 217) and (ii) a control group (n = 105). (i) Adipokines, oxidative stress, indirect autonomic markers, and leucocytes mRNA levels were analysed; (ii) correlation between biomarkers was explored with heatmaps and Kendall correlation coefficients; and (iii) logistic regression and random forest model were used to determine predictors of AF recurrence after ablation. Our results showed that: (i) fatty acid-binding protein 4 (FABP4) and leptin levels were higher in women than in men in both cohorts (P < 0.01). In women, FABP4 levels were higher on AF cohort (20 ± 14 control, 29 ± 18 paroxysmal AF and 31 ± 17 ng/mL persistent AF; P < 0.01). In men, leptin levels were lower on AF cohort (22 ± 15 control, 13 ± 16 paroxysmal AF and 13 ± 11 ng/mL persistent AF; P < 0.01). (ii) In female with paroxysmal AF, there was a lower acetylcholinesterase and higher carbonic anhydrase levels with respect to men (P < 0.05). (iii) Adipokines have an important role on discriminate AF recurrence after ablation. In persistent AF, FABP4 was the best predictor of recurrence after ablation (1.067, 95% confidence interval 1-1.14; P = 0.046). CONCLUSION: The major finding of the present study is the sex-based differences of FABP4 and leptin levels according to AF burden. These adipokines are associated with oxidative stress, inflammatory and autonomic indirect markers, indicating that they may play a role in AF perpetuation.


Subject(s)
Atrial Fibrillation , Catheter Ablation , Fatty Acid-Binding Proteins/blood , Atrial Fibrillation/diagnosis , Atrial Fibrillation/surgery , Female , Humans , Leptin , Male , Recurrence , Sex Characteristics , Treatment Outcome
2.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1500-1509, 2017 06.
Article in English | MEDLINE | ID: mdl-28245984

ABSTRACT

Adverse cardiac remodeling after myocardial infarction (MI) causes impaired ventricular function and heart failure. Histopathological characterization is commonly used to detect the location, size and shape of MI sites. However, the information about chemical composition, physical structure and molecular mobility of peri- and infarct zones post-MI is rather limited. The main objective of this work was to explore the spatiotemporal biochemical and biophysical alterations of key cardiac components post-MI. The FTIR spectra of healthy and remote myocardial tissue shows amides A, I, II and III associated with proteins in freeze-died tissue as major absorptions bands. In infarcted myocardium, the spectrum of these main absorptions was deeply altered. FITR evidenced an increase of the amide A band and the distinct feature of the collagen specific absorption band at 1338cm-1 in the infarct area at 21days post-MI. At 21days post-MI, it also appears an important shift of amide I from 1646cm-1 to 1637cm-1 that suggests the predominance of the triple helical conformation in the proteins. The new spectra bands also indicate an increase in proteoglycans, residues of carbohydrates in proteins and polysaccharides in ischemic areas. Thermal analysis indicates a deep increase of unfreezable water/freezable water in peri- and infarcted tissues. In infarcted tissue is evidenced the impairment of myofibrillar proteins thermal profile and the emergence of a new structure. In conclusion, our results indicate a profound evolution of protein secondary structures in association with collagen deposition and reorganization of water involved in the scar maturation of peri- and infarct zones post-MI.


Subject(s)
Muscle Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Ventricular Remodeling , Animals , Male , Mice , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Protein Structure, Secondary , Spectroscopy, Fourier Transform Infrared/methods
3.
Sci Rep ; 7(1): 47, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28246388

ABSTRACT

Using in vitro, in vivo and patient-based approaches, we investigated the potential of circulating microRNAs (miRNAs) as surrogate biomarkers of myocardial steatosis, a hallmark of diabetic cardiomyopathy. We analysed the cardiomyocyte-enriched miRNA signature in serum from patients with well-controlled type 2 diabetes and with verified absence of structural heart disease or inducible ischemia, and control volunteers of the same age range and BMI (N = 86), in serum from a high-fat diet-fed murine model, and in exosomes from lipid-loaded HL-1 cardiomyocytes. Circulating miR-1 and miR-133a levels were robustly associated with myocardial steatosis in type 2 diabetes patients, independently of confounding factors in both linear and logistic regression analyses (P < 0.050 for all models). Similar to myocardial steatosis, miR-133a levels were increased in type 2 diabetes patients as compared with healthy subjects (P < 0.050). Circulating miR-1 and miR-133a levels were significantly elevated in high-fat diet-fed mice (P < 0.050), which showed higher myocardial steatosis, as compared with control animals. miR-1 and miR-133a levels were higher in exosomes released from lipid-loaded HL-1 cardiomyocytes (P < 0.050). Circulating miR-1 and miR-133a are independent predictors of myocardial steatosis. Our results highlight the value of circulating miRNAs as diagnostic tools for subclinical diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Type 2/blood , Diabetic Cardiomyopathies/blood , MicroRNAs/blood , Myocardium/pathology , Aged , Animals , Biomarkers/blood , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/etiology , Diet, High-Fat , Exosomes , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocytes, Cardiac/pathology , Proton Magnetic Resonance Spectroscopy
4.
Int J Cardiol ; 228: 870-880, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27889554

ABSTRACT

Idiopathic dilated cardiomyopathy has become one of the most prevalent inherited cardiomyopathies over the past decades. Genetic screening of first-degree relatives has revealed that 30-50% of the cases have a familial origin. Similar to other heart diseases, familial dilated cardiomyopathy is characterized by a high genetic heterogeneity that complicates family studies. Cli'nical screening, 12-lead electrocardiogram and transthoracic echocardiogram are recommended for patients and first-degree family members. Magnetic resonance also needs to be considered. Genetic technologies have become fundamental for the clinical management of this disease. New generation sequencing methods have made genetic testing feasible for extensive panels of genes related to the disease. Recently, new imaging modalities such as speckle-tracking, strain and strain rate or magnetic resonance, and circulating biomarkers such as non-coding RNAs, have emerged as potential strategies to help cardiologists in their clinical practice. Imaging, genetic and blood-based techniques should be considered together in the evaluation and testing of familial dilated cardiomyopathy. Here, we discuss the current procedures and novel approaches for the clinical management of familial dilated cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/diagnosis , Biomarkers/blood , Cardiomyopathy, Dilated/blood , Cardiomyopathy, Dilated/genetics , Diagnostic Imaging , Genetic Testing , Humans
5.
Sci Rep ; 6: 37354, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27874027

ABSTRACT

Contractile dysfunction is underdiagnosed in early stages of diabetic cardiomyopathy. We evaluated the potential of circulating long non-coding RNAs (lncRNAs) as biomarkers of subclinical cardiac abnormalities in type 2 diabetes. Forty-eight men with well-controlled type 2 diabetes and 12 healthy age-matched volunteers were enrolled in the study. Left ventricular (LV) parameters were measured by magnetic resonance imaging. A panel of lncRNAs was quantified in serum by RT-qPCR. No differences in expression levels of lncRNAs were observed between type 2 diabetes patients and healthy volunteers. In patients with type 2 diabetes, long intergenic non-coding RNA predicting cardiac remodeling (LIPCAR) was inversely associated with diastolic function, measured as E/A peak flow (P < 0.050 for all linear models). LIPCAR was positively associated with grade I diastolic dysfunction (P < 0.050 for all logistic models). Myocardial infarction-associated transcript (MIAT) and smooth muscle and endothelial cell-enriched migration/differentiation-associated long noncoding RNA (SENCR) were directly associated with LV mass to LV end-diastolic volume ratio, a marker of cardiac remodelling (P < 0.050 for all linear models). These findings were validated in a sample of 30 patients with well-controlled type 2 diabetes. LncRNAs are independent predictors of diastolic function and remodelling in patients with type 2 diabetes.


Subject(s)
Biomarkers/blood , Diabetes Mellitus, Type 2/blood , RNA, Long Noncoding/blood , Ventricular Function, Left , Ventricular Remodeling , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Heart/diagnostic imaging , Heart/physiopathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , RNA, Long Noncoding/genetics
7.
Int J Cardiol ; 201: 20-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26285183

ABSTRACT

BACKGROUND: There is clinical interest in identifying novel lipid biomarkers for evaluating cardiovascular risk and targeting lipid-lowering treatment. Low-density lipoprotein receptor-related protein 1 (LRP1) plays a crucial role in the dysregulated cholesterol transfer from modified lipoproteins to human coronary vascular smooth muscle cells (hVSMCs), promoting hVSMC-derived foam cell formation. LRP1 has a soluble and circulating form (sLRP1) generated from LRP1. Cholesterol modulates the release of the soluble form of LRP1. Using in vitro, ex vivo and patient-based approaches, we tested the association between circulating sLRP1 concentrations and hypercholesterolemia and the potential of sLRP1 as a biomarker of atherosclerosis. METHODS AND RESULTS: Circulating sLRP1 concentrations were higher in severe hypercholesterolemia compared to moderate hypercholesterolemia or normocholesterolemia (Study 1). Circulating sLRP1 was significantly associated with established pro-atherogenic lipid parameters in two different hypercholesterolemic populations (Studies 2 and 3). sLRP1 concentrations decreased after statin treatment and increased after statin withdrawal (Study 3). In vitro experiments showed that native LDL, aggregated LDL and VLDL+IDL lipoproteins induced the release of sLRP1 from hVSMC. sLRP1 levels were increased in the conditioned medium of coronary atherosclerotic plaque areas extracted from patients compared to non-atherosclerotic areas of the same coronary artery and patient. Circulating sLRP1 concentrations were independently associated with the occurrence of carotid atherosclerosis in a hypercholesterolemic population (Study 2). The later association was higher than that observed for other classical or novel lipid parameters. CONCLUSIONS: Circulating sLRP1 is a new lipid-related parameter potentially useful as a biomarker for atherosclerosis.


Subject(s)
Atherosclerosis/blood , Atherosclerosis/diagnosis , Hypercholesterolemia/blood , Hypercholesterolemia/diagnosis , Low Density Lipoprotein Receptor-Related Protein-1/blood , Adult , Aged , Biomarkers/blood , Cells, Cultured , Female , Humans , Male , Middle Aged , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Young Adult
10.
Atherosclerosis ; 233(1): 178-85, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24529141

ABSTRACT

The transcription of the Low-density lipoprotein receptor-related protein (LRP1) is upregulated by aggregated LDL (agLDL) and angiotensin II (AngII) in human vascular smooth muscle cells (hVSMC). The polymorphism c.1-25C>G creates a new GC-box in the LRP1 promoter recognized by Sp1/Sp3 transcription factors. The aims of this study were 1) to evaluate the impact of c.1-25C>G polymorphism on LRP1 transcriptional activity and expression, and 2) to examine the response of c.1-25C>G LRP1 promoter to LDL and AngII. EMSA and Luciferase assays in HeLa cells showed that -25G promoter has enhanced basal transcriptional activity and specific Sp1/Sp3 binding. hVSMC with GG genotype (GG-hVSMC) had higher LRP1 mRNA and protein levels, respectively than CC genotype (CC-hVSMC). EMSA assays showed that the polymorphism determines scarce amount of SRE-B/SREBP-2 complex formation and the failure of agLDL to further reduce these SRE-B/SREBP-2 complexes. Taken together, these results suggest that c.1-25C>G, by difficulting SREBP-2 binding, prevents SREBP-2 displacement required for LRP1 promoter response to LDL. In contrast, c.1-25C>G strongly favours Sp1/Sp3 binding and AngII-induced activity in Sp1/Sp3 dependent manner in GG-hVSMC. This increase is functionally translated into a higher capacity of GG-hVSMC to become foam cells from agLDL in presence of AngII. These results suggest that c.1-25C>G determines a lack of response to agLDL and an exacerbated response to AngII. It is thus conceivable that the presence of the polymorphism would be easily translated to vascular alterations in the presence of the pro-hypertensive autacoid, AngII.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/genetics , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Angiotensin II/physiology , Binding Sites , HeLa Cells , Humans , Lipoproteins, LDL/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/biosynthesis , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Polymorphism, Genetic , Sterol Regulatory Element Binding Protein 2/biosynthesis , Transcriptional Activation
11.
Acta Diabetol ; 51(1): 23-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23096408

ABSTRACT

Lipoprotein receptor expression plays a crucial role in the pathophysiology of adipose tissue in in vivo models of diabetes. However, there are no studies in diabetic patients. The aims of this study were to analyze (a) low-density lipoprotein receptor-related protein 1 (LRP1) and very low-density lipoprotein receptor (VLDLR) expression in epicardial and subcutaneous fat from type 2 diabetes mellitus compared with nondiabetic patients and (b) the possible correlation between the expression of these receptors and plasmatic parameters. Adipose tissue biopsy samples were obtained from diabetic (n = 54) and nondiabetic patients (n = 22) undergoing cardiac surgery before the initiation of cardiopulmonary bypass. Adipose LRP1 and VLDLR expression was analyzed at mRNA level by real-time PCR and at protein level by Western blot analysis. Adipose samples were also subjected to lipid extraction, and fat cholesterol ester, triglyceride, and free cholesterol contents were analyzed by thin-layer chromatography. LRP1 expression was higher in epicardial fat from diabetic compared with nondiabetic patients (mRNA 17.63 ± 11.37 versus 7.01 ± 4.86; P = 0.02; protein 11.23 ± 7.23 versus 6.75 ± 5.02, P = 0.04). VLDLR expression was also higher in epicardial fat from diabetic patients but only at mRNA level (231.25 ± 207.57 versus 56.64 ± 45.64, P = 0.02). No differences were found in the expression of LRP1 or VLDLR in the subcutaneous fat from diabetic compared with nondiabetic patients. Epicardial LRP1 and VLDLR mRNA overexpression positively correlated with plasma triglyceride levels (R(2) = 0.50, P = 0.01 and R(2) = 0.44, P = 0.03, respectively) and epicardial LRP1 also correlated with plasma glucose levels (R(2) = 0.33, P = 0.03). These results suggest that epicardial overexpression of certain lipoprotein receptors such as LRP1 and VLDLR expression may play a key role in the alterations of lipid metabolism associated with type 2 diabetes mellitus.


Subject(s)
Adipose Tissue/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Pericardium/metabolism , Triglycerides/blood , Aged , Case-Control Studies , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Lipid Metabolism/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Male , Middle Aged , Receptors, LDL/genetics , Receptors, LDL/metabolism , Subcutaneous Fat/metabolism , Up-Regulation
12.
Atherosclerosis ; 227(1): 79-88, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23312784

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) negatively modulate the expression of the CD91/low-density lipoprotein receptor-related protein (LRP1), a carrier and signaling receptor that mediates the endocytosis of more than 40 structurally and functionally distinct ligands. The aim of this work was to analyze whether lipopolysaccharide (LPS) can regulate LRP1 expression through SREBPs in human monocyte-derived macrophages (HMDM). LPS led to LRP1 mRNA and protein inhibition in a dose- and time-dependent manner. Concomitantly, a strong upregulation of SREBP-1 mRNA and SREBP-1 nuclear protein levels was observed in LPS-treated HMDM. The specific silencing of SREBP-1 efficiently prevented LRP1 reduction caused by LPS. SREBP-1 mRNA and nuclear protein levels remained high in HMDM treated with LPS unexposed or exposed to LDL. Native (nLDL) or aggregated LDL (agLDL) per se downregulated SREBP-2 expression levels and increased LRP1 expression. However, lipoproteins did not significantly alter the effect of LPS on SREBP-1 and LRP1 expression. Collectively, these data support that lipoproteins and LPS exert their modulatory effect on LRP1 expression through different SREBP isoforms, SREBP-2 and SREBP-1, respectively. These results highlight a crucial role of SREBP-1 as a mediator of the downregulatory effects of LPS on LRP1 expression in human macrophages, independently of the absence or presence of modified lipoproteins.


Subject(s)
Lipopolysaccharides/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/biosynthesis , Macrophages/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Down-Regulation , Humans , Macrophages/drug effects , Nuclear Proteins/metabolism , RNA, Messenger/metabolism
13.
Nutr Metab Cardiovasc Dis ; 23(3): 257-63, 2013 Mar.
Article in English | MEDLINE | ID: mdl-21824756

ABSTRACT

BACKGROUND AND AIMS: Epidemiological studies have demonstrated an association between high-polyphenol intake and reduced incidence of atherosclerosis. The healthy effects of cocoa-polyphenols may be due to their antioxidant and anti-inflammatory actions, although the exact mechanisms are unknown and depend on the matrix in which cocoa-polyphenols are delivered. Nuclear factor κB (NF-κB) is a key molecule in the pathophysiology of atherosclerosis involved in the regulation of adhesion molecules(AM) and cytokine expression and its activation is the first step in triggering the inflammatory process. The aim of this study was to evaluate the effect of acute cocoa consumption in different matrices related to the bioavailability of cocoa-polyphenols in NF-κB activation and the expression of AM. METHODS AND RESULTS: Eighteen healthy volunteers randomly received 3 interventions: 40g of cocoa powder with milk (CM), with water (CW), and only milk (M). NF-κB activation in leukocytes and AM (sICAM, sVCAM, E-selectin) were measured before and 6h after each intervention. Consumption of CW significantly decreased NF-κB activation compared to baseline and to CM (P < 0.05, both), did not change after CM intervention, and significantly increased after M intervention (P = 0.014). sICAM-1 concentrations significantly decreased after 6h of CW and CM interventions (P ≤ 0.026; both) and E-selectin only decreased after CW intervention (P = 0.028). No significant changes were observed in sVCAM-1 concentrations. CONCLUSIONS: The anti-inflammatory effect of cocoa intake may depend on the bioavailability of bioactive compounds and may be mediated at least in part by the modulation of NF-κB activation and downstream molecules reinforcing the link between cocoa intake and health.


Subject(s)
Beverages , Cacao/chemistry , Leukocytes, Mononuclear/drug effects , Adult , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Biological Availability , Blotting, Western , Cell Adhesion Molecules , Cross-Over Studies , E-Selectin/genetics , E-Selectin/metabolism , Female , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Male , Middle Aged , Milk , NF-kappa B , Polyphenols/administration & dosage , Polyphenols/pharmacokinetics , Prospective Studies , Signal Transduction , Transcription Factors , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
14.
Acta Diabetol ; 50(4): 553-62, 2013 Aug.
Article in English | MEDLINE | ID: mdl-21997325

ABSTRACT

Inflammatory markers are elevated in type 2 diabetic patients (DP) and may predict the development of type 2 diabetes. Our aims were to analyze differences in the expression of inflammatory and immunological molecules between DP and healthy subjects and to investigate whether glycemic control might prevent the overexpression of inflammatory markers in DP. Twenty-two DP with advanced atherosclerosis and eight healthy blood donors were included. DP were classified as well (HbA1c ≤ 6.5) or poorly controlled (HbA1c > 6.5). In "in vitro" studies, monocytes were exposed to low (5.5 mM) or high glucose (26 mM) concentrations in the absence or presence of insulin. Expression profiling of 14 inflammatory genes was analyzed using TLDA analysis. "In vivo" results show that monocytes from DP had increased levels of monocyte chemoattractant protein (MCP-1) and interleukin 6 (IL6) and lower levels of Toll-like receptor 2 (TLR2) mRNA than healthy subjects. Well-controlled DP had lower levels of IL-6 than poorly controlled DP, suggesting that glycemic control may prevent IL6 mRNA alterations associated with diabetes. "In vitro" results demonstrate that glucose directly and significantly induced MCP-1 and IL6 and reduced TLR2 mRNA expression. Insulin at high dose (100 IU/ml) dramatically enhanced the upregulatory effects of glucose on MCP-1 and IL-6 and reduced per se TLR2 mRNA expression. MCP-1, IL-6 and TLR2 are key inflammatory players altered in monocytes from type 2 DP. Both hyperinsulinemia and hyperglycemia contribute to alter the expression of these genes. The glycemic control only significantly prevented IL6 overexpression in this group of patients.


Subject(s)
Atherosclerosis/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/genetics , Inflammation Mediators/metabolism , Monocytes/metabolism , Aged , Atherosclerosis/etiology , Atherosclerosis/metabolism , Biomarkers/blood , Biomarkers/metabolism , Case-Control Studies , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Female , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Middle Aged , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
15.
J Cell Physiol ; 227(10): 3528-33, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22392894

ABSTRACT

Human hepatic stellate cells (HHSCs) proliferation and migration play a key role in the pathogenesis of liver inflammation and fibrogenesis. Low density lipoprotein receptor-related protein (LRP1) is an endocytic receptor involved in intracellular signal transduction. The aim of this work was to analyse the role of low density lipoprotein receptor-related protein (LRP1) in HHSCs proliferation and migration and the mechanisms involved. Human LRP1 deficient-HHSCs were generated by nucleofecting the line HHSCs with siRNA anti-LRP1. HHSCs DNA synthesis was measured by [(3) H]-thymidine incorporation and cell cycle progression by flow cytometry after annexin V and iodure propidium staining. Cell migration was assessed using a wound repair model system. LRP1 expression and extracellular matrix-regulated kinase (ERK1,2) phosphorylation were analysed by Western blot analysis. Transforming growth factor-ß (TGF-ß) extracellular levels were analysed by ELISA. siRNA-antiLRP1 treatment almost completely inhibited LRP1 mRNA and protein expression. LRP1 deficient HHSCs showed higher proliferative response (172 ± 19 vs. 93 ± 8 [(3) H]-thymidine incorporation; 78.68% vs. 82.69% in G0/G1, 21.32% vs. 17.30% in G2/S) and higher migration rates than control HHSCs. LRP1 deficient cells showed higher levels of phosphorylated ERK1,2. TGF-ß extracellular levels were threefold higher in LRP1-deficient than in control HHSCs cells. These results demonstrate that LRP1 regulates HHSCs proliferation and migration through modulation of ERK1,2 phosphorylation and TGF-ß extracellular levels. These results suggest that HHSCs-LRP1 may play a key role in the modulation of factors determining hepatic fibrosis.


Subject(s)
Cell Movement/genetics , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Cell Cycle/genetics , Cell Growth Processes/genetics , Cells, Cultured , Humans , MAP Kinase Signaling System/genetics , Phosphorylation , RNA, Messenger/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
16.
Atherosclerosis ; 213(2): 458-68, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980003

ABSTRACT

Low density lipoprotein receptor-related protein (LRP1) is upregulated in vascular smooth muscle cells by intravascular aggregated LDL (agLDL) - LDL trapped in the arterial intima and systemic LDL. LRP1 upregulation in hypercholesterolemic aortas is concomitant with SREBP downregulation. However, the specific role of SREBP isoforms in LRP1 transcription and LDL-induced LRP1 upregulation in human vascular smooth muscle cells (VSMC) is unknown. In the present study we report that specific silencing of either SREBP-1 or SREBP-2 enhanced LRP1 whereas overexpression of the active SREBP isoforms decreased LRP1 expression. Gel mobility shift and ChIP assays demonstrated that SREBP-1a, SREBP-1c and SREBP-2 were able to bind to three putative SRE sequences; SRE-A (-1042 to -1028), SRE-B (-115 to -101) and SRE-C (+226 to +234). ChIP assays demonstrated that agLDL (100µg/mL, 24h) significantly and specifically decreased SREBP-2 binding to the LRP1 promoter. Luciferase assays demonstrated that agLDL increased the transcriptional activity of A/B or A/C double mutants but failed to increase that of the double B/C mutant. Our results show that both SREBP-1 and SREBP-2 negatively modulated LRP1 transcription. Furthermore, agLDL exerted an upregulatory effect on LRP1 expression by decreasing SREBP-2 binding to LRP1 promoter. Two SRE-like sequences control the response of LRP1 to agLDL.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/biosynthesis , Muscle, Smooth, Vascular/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Down-Regulation , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Protein Isoforms/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Up-Regulation
17.
J Thromb Haemost ; 7(12): 2137-46, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19817993

ABSTRACT

BACKGROUND: Our previous results demonstrated that aggregated low density lipoprotein (agLDL) induces tissue factor (TF) expression and activation through Rho A translocation in human vascular smooth muscle cells (VSMC). We also previously demonstrated that membrane sphingomyelin (SM) content is higher in agLDL-exposed VSMC than in control cells. The main enzymes regulating cellular SM content are the family of sphingomyelinases (Smases) that hydrolize SM to phosphorylcholine and ceramide (CER). OBJECTIVES: We wished to investigate whether agLDL has the ability to modulate acidic- (A-) and neutral (N-) Smase activity and whether or not this effect is related to the upregulatory effect of agLDL on Rho A translocation and TF activation in human VSMC. METHODS AND RESULTS: By measuring generated [(14)C]-phosphorylcholine, we found that agLDL significantly decreased A-Smase and specially N-Smase activity. Pharmacological Smase inhibitors increased Rho A and TF. Specific loss-of-function of A-Smase or N-Smase 1 (N1-Smase) by siRNA treatment (500 nmol L(-1), 12 hours) dramatically increased membrane Rho A protein levels (5- and 3-fold, respectively). Concomitantly, TF protein expression and TF procoagulant activity were also increased. Inhibition of A-Smase or N-Smase activity by agLDL, siRNA-anti A- or N1-Smase or pharmacological treatment significantly increased the SM content of vascular cells. The inhibition of SM synthesis by fumonisin B(1) (FB(1)) prevented the upregulatory effect of agLDL on TF. CONCLUSIONS: These results demonstrate that inhibition of both A- and N1-Smase might explain the upregulatory effect of agLDL on TF activation, and suggest that this effect is related, at least in part, to membrane SM enrichment.


Subject(s)
Lipoproteins, LDL/physiology , Muscle, Smooth, Vascular/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Thromboplastin/biosynthesis , Up-Regulation/genetics , Humans , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Protein Multimerization , Protein Transport , rho-Associated Kinases/metabolism
18.
Trauma (Majadahonda) ; 19(3): 152-159, jul.-sept. 2008. tab, ilus
Article in Spanish | IBECS | ID: ibc-84394

ABSTRACT

Objetivo: Analizar si la unión e interiorización de la LDL modificada por agregación (LDLag) puede inducir la expresión de la proteina adipofilina (ADRP), un marcador de acumulación lipídica, en las células musculares lisas de la pared vascular (CMLV) y macrófagos humanos. Resultados: La LDLag induce la sobreexpresión de ADRP tanto a nivel de ARNm (PCR tiempo real) como a nivel de proteina («western blot») en CMLV (ARNm: 3.06-veces; proteina: 8.58-veces) y también en macrófagos (ARNm: 3.5-veces; proteina: 3.71-veces). Los estudios immunohistoquímicos evidenciaron una alta colocalización entre ADRP y CMLV y también entre ADRP y macrófagos en placas ateroscleróticas avanzadas ricas en lípido. Conclusiones: La captación de LDLag mediante el receptor lipoproteíco LRP1 juega un papel primordial en la formación de células espumosas a partir de macrófagos y de CMLV y, por tanto, en la progresión de la lesión aterosclerótica (AU)


Aims: The objectives of this work were to analyze whether aggregated LDL (agLDL) uptake modulates ADRP expression levels in human vascular smooth muscle cells (VSMC) and macrophages (HMDM). Methods and Results: AgLDL strongly upregulated ADRP mRNA (Real-time PCR) and protein expression (western blot) in human VSMC (mRNA: by 3.06-fold; protein: 8.58-fold) and HMDM (mRNA: by 3.5-fold; protein: by 3.71-fold).. Immunohystochemical studies evidence a high colocolocalization between ADRP/macrophages and ADRP/VSMC in advanced lipid-enriched atherosclerotic plaques. Conclusions:These results demonstrate that agLDL-LRP1 engagement induces ADRP overexpression in both HMDM and human VSMC. ADRP is highly expressed in advanced lipid-enriched human atherosclerotic plaques. Therefore, LRP1-mediated agLDL uptake might play a pivotal role on vascular foam cell formation and atherosclerotic plaque progression (AU)


Subject(s)
Humans , Male , Female , Receptors, LDL/administration & dosage , Receptors, LDL/analysis , Cholesterol, LDL/analysis , Lipoproteins, LDL/analysis , Arteriosclerosis/complications , Arteriosclerosis/diagnosis , Polymerase Chain Reaction/methods , Macrophages/metabolism , Muscle Cells/metabolism , Arteriosclerosis/etiology , Myocytes, Smooth Muscle , Myocytes, Smooth Muscle/metabolism , Immunohistochemistry/methods
19.
J Lipid Res ; 48(10): 2133-40, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17620659

ABSTRACT

Aggregated LDL (agLDL) is internalized by LDL receptor-related protein (LRP1) in vascular smooth muscle cells (VSMCs) and human monocyte-derived macrophages (HMDMs). AgLDL is, therefore, a potent inducer of massive intracellular cholesteryl ester accumulation in lipid droplets. The adipocyte differentiation-related protein (ADRP) has been found on the surface of lipid droplets. The objectives of this work were to analyze whether agLDL uptake modulates ADRP expression levels and whether the effect of agLDL internalization on ADRP expression depends on LRP1 in human VSMCs and HMDMs. AgLDL strongly upregulates ADRP mRNA (real-time PCR) and protein expression (Western blot) in human VSMCs (mRNA: by 3.06-fold; protein: 8.58-fold) and HMDMs (mRNA: by 3.5-fold; protein: by 3.71-fold). Treatment of VSMCs and HMDMs with small anti-LRP1-interfering RNA (siRNA-LRP1) leads to specific inhibition of LRP1 expression. siRNA-LRP1 treatment significantly reduced agLDL-induced ADRP overexpression in HMDMs (by 69%) and in VSMCs (by 53%). Immunohystochemical studies evidence a colocolocalization between ADRP/macrophages and ADRP/VSMCs in advanced lipid-enriched atherosclerotic plaques. These results demonstrate that agLDL-LRP1 engagement induces ADRP overexpression in both HMDMs and human VSMCs and that ADRP is highly expressed in advanced lipid-enriched human atherosclerotic plaques. Therefore, LRP1-mediated agLDL uptake might play a pivotal role in vascular foam cell formation.


Subject(s)
Adipocytes/metabolism , Lipoproteins, LDL/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , CD36 Antigens/biosynthesis , Cell Differentiation , Cholesterol/metabolism , Foam Cells/metabolism , Humans , Immunohistochemistry/methods , Lipids/chemistry , RNA/metabolism , RNA, Messenger/metabolism
20.
Cardiovasc Res ; 73(1): 208-16, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17141748

ABSTRACT

OBJECTIVE: Low density lipoprotein (LDL) internalized in the vascular wall and modified by binding to extracellular matrix-proteoglycans (ECM) becomes aggregated (agLDL). AgLDL induces tissue factor (TF) expression and activity in human vascular smooth muscle cells (VSMC). TF expression in vascular cells promotes the prothrombotic transformation of the vascular wall. However, the mechanisms by which agLDL induces TF are not known. The aim of this study was to investigate the mechanisms involved in TF activation by extracellular matrix-modified LDL in human VSMC. METHODS AND RESULTS: AgLDL significantly induces TF expression (real time PCR and Western blot analysis) and procoagulant activity (factor Xa generation test) in human VSMC. HMG-CoA reductase inhibition completely prevents agLDL-induced TF expression and partially inhibits agLDL-TF activation. These effects are reverted by geranylgeranyl pyrophosphate (GGPP) but not by farnesyl pyrophosphate (FPP), suggesting the involvement of a geranylated protein in agLDL-TF induction. AgLDL increases Rho A translocation (2-fold) from the cytoplasm to the cell membrane in control but not in simvastatin-treated VSMC. Exoenzyme C3, a specific Rho A inhibitor, completely prevents agLDL-induced TF overexpression and partially agLDL-TF activation. Blocking LRP1, the receptor of agLDL, with anti-LRP1 antibodies or inhibiting LRP1 expression by small interference RNA treatment (siRNA-LRP1) impairs agLDL-induced TF overexpression and activation. CONCLUSIONS: These results demonstrate that TF induction by agLDL depends on LRP1 expression and requires Rho A translocation to the cellular membrane.


Subject(s)
Lipoproteins, LDL/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/metabolism , Thromboplastin/metabolism , rhoA GTP-Binding Protein/metabolism , ADP Ribose Transferases/pharmacology , Antibodies, Monoclonal/pharmacology , Biological Transport , Blotting, Western/methods , Botulinum Toxins/pharmacology , Cell Membrane/metabolism , Cells, Cultured , Coronary Vessels , Cytoplasm/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunohistochemistry/methods , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/immunology , Polyisoprenyl Phosphates/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sesquiterpenes/pharmacology , Simvastatin/pharmacology , Thromboplastin/analysis , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...