Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Eur J Prev Cardiol ; 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861400

ABSTRACT

AIM: To assess whether implementation of the 2019 ESC/EAS dyslipidaemia guidelines observed between 2020-2021 improved between 2021-2022 in the SANTORINI study. METHODS: High- or very-high cardiovascular (CV) risk patients were recruited across 14 European countries from March 2020-February 2021, with 1-year prospective follow-up until May 2022. Lipid-lowering therapy (LLT) and 2019 ESC/EAS risk-based low-density lipoprotein cholesterol (LDL-C) goal attainment (defined as <1.4 mmol/L for patients at very high CV risk and <1.8 mmol/L for patients at high CV risk) at 1-year follow-up were compared with baseline. . RESULTS: Of 9559 patients enrolled, 9136 (2626 high risk, 6504 very high risk) had any follow-up data, and 7210 (2033 high risk, 5173 very high risk) had baseline and follow-up LDL-C data. LLT was escalated in one-third of patients and unchanged in two-thirds. Monotherapy and combination therapy usage rose from 53.6% and 25.6% to 57.1% and 37.9%, respectively. Mean LDL-C levels decreased from 2.4 mmol/L to 2.0 mmol/L. Goal attainment improved from 21.2% to 30.9%, largely driven by LLT use among those not on LLT at baseline. Goal attainment was greater with combination therapy compared with monotherapy at follow-up (39.4 vs 25.5%). CONCLUSIONS: LLT use and achievement of risk-based lipid goals increased over 1-year follow-up particularly when combination LLT was used. Nonetheless, most patients remained above goal, hence strategies are needed to improve implementation of combination LLT.


Cardiovascular diseases, a group of disorders of the heart and blood vessels, are the most common cause of death worldwide. Lowering low-density lipoprotein (LDL) cholesterol in the bloodstream reduces the risk of developing cardiovascular diseases, such as heart attacks and strokes. Guidelines recommend that those at highest risk of cardiovascular disease should achieve the lowest levels of LDL cholesterol. Several medications are available that help lower LDL cholesterol levels and prevent cardiovascular events, however, recent studies have shown that the majority of patients continue to have LDL cholesterol levels above optimal value in part due to suboptimal use of these medications. Here we report the results after 1 year of follow-up of the SANTORINI study (started in 2020) which aimed to document the management of LDL cholesterol in clinical practice across 14 countries in Europe. We found that better control of LDL cholesterol occurred when more than one drug was used (combination therapy). Use of combination therapy was low at the start of the study 25.6% but increased over 1 year to 37.9%, resulting in better control of LDL cholesterol at 1 year than observed at the start of the study. Nonetheless, only 31% of patients achieved their LDL cholesterol target levels based on the European guidelines. Greater use of combination therapies is needed in order to improve the overall population level control of LDL cholesterol.

2.
Curr Med Res Opin ; 37(12): 2049-2059, 2021 12.
Article in English | MEDLINE | ID: mdl-34517739

ABSTRACT

OBJECTIVE: To provide a contemporary overview of recent real-world lipid-lowering therapy (LLT) practices and outcomes in patients with hypercholesterolemia/dyslipidemia at high/very high risk of atherosclerotic cardiovascular disease in Europe. METHODS: A structured literature review of recent (July 2015-July 2020) real-world studies reporting lipid management and outcomes was conducted using a rapid evidence synthesis. Outcomes included patient characteristics, LLT treatment practices, adherence and low-density lipoprotein cholesterol (LDL-C) goal attainment. RESULTS: Fifty-three real-world observational studies in high/very high risk patients were selected after screening 5664 records (n = 50 national [sample size range 38-237,279] and n = 3 multinational studies [sample size range 6648-8456]). Mean age ranged from 33 to 77 years; hypertension, diabetes and obesity were commonly reported comorbidities. Statins were the most common LLT; patients without familial hypercholesterolemia (FH) mostly received high or moderate intensity statins/LLT, while patients with FH mostly received high intensity statins/LLT. The proportion of patients receiving ezetimibe was low overall (ezetimibe + statin use in those with and without familial hypercholesterolemia [FH] range 5%-59% and 1%-22%, respectively). Overall, the use of proprotein convertase subtilisin/kexin 9 inhibitor (PCSK9i) therapy was limited. Adherence to LLT therapies was defined variably and ranged from 46%-92%. LDL-C goal attainment was suboptimal, irrespective of LLT (overall range in goal attainment with oral LLT was 2%-73% [FH: 2%-23%] and with PCSK9i was 20%-65%). CONCLUSIONS: LDL-C control is suboptimal and the available LLT armamentarium, most importantly combination therapy, is being underutilized in high/very high risk patients leading to inadequate management of cardiovascular risk.


Subject(s)
Anticholesteremic Agents , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hyperlipoproteinemia Type II , Adult , Aged , Cholesterol, LDL , Europe , Ezetimibe , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Lipids , Middle Aged , Treatment Outcome
3.
Sci Rep ; 11(1): 16419, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385562

ABSTRACT

Total body upstream stimulatory factor 1 (USF1) deficiency in mice is associated with brown adipose tissue activation and a marked protection against the development of obesity and atherosclerotic lesions. Functional expression of USF1 has also been detected in monocytes and monocyte-derived macrophages. In the current study we therefore tested whether selective hematopoietic USF1 deficiency can also beneficially impact the development of atherosclerosis. For this purpose, LDL receptor knockout mice were transplanted with bone marrow from USF1 knockout mice or their wild-type littermate controls and subsequently fed a Western-type diet for 20 weeks to stimulate atherosclerotic lesion development. Strikingly, absence of USF1 function in bone marrow-derived cells was associated with exacerbated blood leukocyte (+ 100%; P < 0.01) and peritoneal leukocyte (+ 50%; P < 0.05) lipid loading and an increased atherosclerosis susceptibility (+ 31%; P < 0.05). These effects could be attributed to aggravated hyperlipidemia, i.e. higher plasma free cholesterol (+ 33%; P < 0.001) and cholesteryl esters (+ 39%; P < 0.001), and the development of hepatosteatosis. In conclusion, we have shown that hematopoietic USF1 deficiency is associated with an increased atherosclerosis susceptibility in LDL receptor knockout mice. These findings argue against a contribution of macrophage-specific USF1 deficiency to the previously described beneficial effect of total body USF1 deficiency on atherosclerosis susceptibility in mice.


Subject(s)
Atherosclerosis/genetics , Genetic Predisposition to Disease , Receptors, LDL/genetics , Upstream Stimulatory Factors/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Lipids Health Dis ; 17(1): 285, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30545366

ABSTRACT

BACKGROUND: The focus of studies on high-density lipoproteins (HDL) has shifted from HDL-cholesterol (HDL-C) to HDL function. We recently demonstrated that low USF1 expression in mice and humans associates with high plasma HDL-C and low triglyceride levels, as well as protection against obesity, insulin resistance, and atherosclerosis. Here, we studied the impact of USF1 deficiency on HDL functional capacity and macrophage atherogenic functions, including inflammation, cholesterol efflux, and cholesterol accumulation. METHODS: We used a congenic Usf1 deficient mice in C57Bl/6JRccHsd background and blood samples were collected to isolate HDL for structural and functional studies. Lentiviral preparations containing the USF1 silencing shRNA expression vector were used to silence USF1 in human THP-1 and Huh-7 cells. Cholesterol efflux from acetyl-LDL loaded THP-1 macrophages was measured using HDL and plasma as acceptors. Gene expression analysis from USF1 silenced peritoneal macrophages was carried out using Affymetrix protocols. RESULTS: We show that Usf1 deficiency not only increases HDL-C levels in vivo, consistent with elevated ABCA1 protein expression in hepatic cell lines, but also improves the functional capacity of HDL particles. HDL particles derived from Usf1 deficient mice remove cholesterol more efficiently from macrophages, attributed to their higher contents of phospholipids. Furthermore, silencing of USF1 in macrophages enhanced the cholesterol efflux capacity of these cells. These findings are consistent with reduced inflammatory burden of USF1 deficient macrophages, manifested by reduced secretion of pro-inflammatory cytokines MCP-1 and IL-1ß and protection against inflammation-induced macrophage cholesterol accumulation in a cell-autonomous manner. CONCLUSIONS: Our findings identify USF1 as a novel factor regulating HDL functionality, showing that USF1 inactivation boosts cholesterol efflux, reduces macrophage inflammation and attenuates macrophage cholesterol accumulation, linking improved macrophage cholesterol metabolism and inflammatory pathways to the antiatherogenic function of USF1 deficiency.


Subject(s)
Cholesterol, HDL/genetics , Cholesterol/genetics , Lipoproteins, HDL/genetics , Upstream Stimulatory Factors/genetics , ATP Binding Cassette Transporter 1/genetics , Animals , Chemokine CCL2/genetics , Cholesterol/blood , Gene Expression/genetics , Humans , Inflammation/blood , Inflammation/genetics , Inflammation/pathology , Insulin Resistance/genetics , Lipoproteins, HDL/blood , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Knockout , Obesity/blood , Obesity/genetics , Obesity/pathology
5.
Sci Transl Med ; 8(323): 323ra13, 2016 Jan 27.
Article in English | MEDLINE | ID: mdl-26819196

ABSTRACT

USF1 (upstream stimulatory factor 1) is a transcription factor associated with familial combined hyperlipidemia and coronary artery disease in humans. However, whether USF1 is beneficial or detrimental to cardiometabolic health has not been addressed. By inactivating USF1 in mice, we demonstrate protection against diet-induced dyslipidemia, obesity, insulin resistance, hepatic steatosis, and atherosclerosis. The favorable plasma lipid profile, including increased high-density lipoprotein cholesterol and decreased triglycerides, was coupled with increased energy expenditure due to activation of brown adipose tissue (BAT). Usf1 inactivation directs triglycerides from the circulation to BAT for combustion via a lipoprotein lipase-dependent mechanism, thus enhancing plasma triglyceride clearance. Mice lacking Usf1 displayed increased BAT-facilitated, diet-induced thermogenesis with up-regulation of mitochondrial respiratory chain complexes, as well as increased BAT activity even at thermoneutrality and after BAT sympathectomy. A direct effect of USF1 on BAT activation was demonstrated by an amplified adrenergic response in brown adipocytes after Usf1 silencing, and by augmented norepinephrine-induced thermogenesis in mice lacking Usf1. In humans, individuals carrying SNP (single-nucleotide polymorphism) alleles that reduced USF1 mRNA expression also displayed a beneficial cardiometabolic profile, featuring improved insulin sensitivity, a favorable lipid profile, and reduced atherosclerosis. Our findings identify a new molecular link between lipid metabolism and energy expenditure, and point to the potential of USF1 as a therapeutic target for cardiometabolic disease.


Subject(s)
Adipose Tissue, Brown/metabolism , Upstream Stimulatory Factors/deficiency , Upstream Stimulatory Factors/genetics , Adult , Aged , Alleles , Animals , Atherosclerosis/metabolism , Blood Glucose/metabolism , Carbohydrates/chemistry , Cardiovascular System , Cholesterol, HDL/blood , Cholesterol, HDL/metabolism , Cohort Studies , Female , Gene Silencing , Glucose/metabolism , Humans , Insulin/blood , Insulin/metabolism , Lipids/chemistry , Lipoprotein Lipase/metabolism , Lipoproteins, VLDL/metabolism , Liver/metabolism , Male , Metabolic Syndrome/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Oxygen Consumption , Phenotype , Polymorphism, Single Nucleotide , Thermogenesis , Triglycerides/blood , Triglycerides/metabolism
6.
Biochim Biophys Acta ; 1861(4): 342-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26747651

ABSTRACT

We investigated the expression of miR-192* (miR-192-3p) in the visceral adipose tissue (VAT) of obese subjects and its function in cultured human adipocytes. This miRNA is a 3' arm derived from the same pre-miRNA as miR-192 (miR-192-5p) implicated in type 2 diabetes, liver disease and cancers, and is predicted to target key genes in lipid metabolism. In morbidly obese subjects undergoing bariatric surgery preceded by a very low calorie diet, miR-192* in VAT correlated negatively (r=-0.387; p=0.046) with serum triglyceride (TG) and positively with high-density lipoprotein (HDL) concentration (r=0.396; p=0.041). In a less obese patient cohort, the miRNA correlated negatively with the body mass index (r=-0.537; p=0.026). To characterize the function of miR-192*, we overexpressed it in cultured adipocytes and analyzed the expression of adipogenic differentiation markers as well as cellular TG content. Reduced TG and expression of the adipocyte marker proteins aP2 (adipocyte protein 2) and perilipin 1 were observed. The function of miR-192* was further investigated by transcriptomic profiling of adipocytes expressing this miRNA, revealing impacts on key lipogenic genes. A number of the mRNA alterations were validated by qPCR. Western analysis confirmed a marked reduction of the lipogenic enzyme SCD (stearoyl coenzyme A desaturase-1), the fatty aldehyde dehydrogenase ALDH3A2 (aldehyde dehydrogenase 3 family member A2) and the high-density lipoprotein receptor SCARB1 (scavenger receptor B, type I). SCD and ALDH3A2 were demonstrated to be direct targets of miR-192*. To conclude, the present data identify miR-192* as a novel controller of adipocyte differentiation and lipid homeostasis.


Subject(s)
Adipocytes, White/metabolism , Adipogenesis , Intra-Abdominal Fat/metabolism , MicroRNAs/metabolism , Obesity, Morbid/metabolism , Triglycerides/metabolism , Adipogenesis/genetics , Adult , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Body Mass Index , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Lipoproteins, HDL/blood , Male , MicroRNAs/genetics , Middle Aged , Obesity, Morbid/blood , Obesity, Morbid/diagnosis , Obesity, Morbid/genetics , Perilipin-1 , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Messenger/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , Transfection , Triglycerides/blood
7.
Physiol Rep ; 4(1)2016 Jan.
Article in English | MEDLINE | ID: mdl-26733244

ABSTRACT

MicroRNAs (miRNAs) control gene expression by reducing mRNA stability and translation. We aimed to identify alterations in human liver miRNA expression/function in nonalcoholic fatty liver disease (NAFLD). Subjects with the highest (median liver fat 30%, n = 15) and lowest (0%, n = 15) liver fat content were selected from >100 obese patients for miRNA profiling of liver biopsies on microarrays carrying probes for 1438 human miRNAs (a cross-sectional study). Target mRNAs and pathways were predicted for the miRNAs most significantly upregulated in NAFLD, their cell-type-specific expression was investigated by quantitative PCR (qPCR), and the transcriptome of immortalized human hepatocytes (IHH) transfected with the miRNA with the highest number of predicted targets, miR-576-5p, was studied. The screen revealed 42 miRNAs up- and two downregulated in the NAFLD as compared to non-NAFLD liver. The miRNAs differing most significantly between the groups, miR-103a-2*, miR-106b, miR-576-5p, miRPlus-I137*, miR-892a, miR-1282, miR-3663-5p, and miR-3924, were all upregulated in NAFLD liver. Target pathways predicted for these miRNAs included ones involved in cancer, metabolic regulation, insulin signaling, and inflammation. Consistent transcriptome changes were observed in IHH transfected with miR-576-5p, and western analysis revealed a marked reduction of the RAC1 protein belonging to several miR-576-5p target pathways. To conclude, we identified 44 miRNAs differentially expressed in NAFLD versus non-NAFLD liver, 42 of these being novel in the context of NAFLD. The study demonstrates that by applying a novel study set-up and a broad-coverage array platform one can reveal a wealth of previously undiscovered miRNA dysregulation in metabolic disease.


Subject(s)
Liver/metabolism , MicroRNAs/biosynthesis , Non-alcoholic Fatty Liver Disease/metabolism , Up-Regulation/physiology , Adult , Cell Line, Transformed , Cells, Cultured , Cross-Sectional Studies , Female , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/pathology , Liver/surgery , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/surgery
8.
J Clin Endocrinol Metab ; 100(10): E1299-307, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26204133

ABSTRACT

OBJECTIVE: Circulating ANGPTL8 has recently been used as a marker of insulin action. We studied expression and insulin regulation of ANGPTL8 and ANGPTL3 in vivo and in vitro. DESIGN AND METHODS: Expression of ANGPTL8 and ANGPTL3 was studied in 34 paired samples of human liver and adipose tissue. Effects of insulin on 1) plasma concentrations and adipose tissue expression of ANGPTL8 and ANGPTL3 (in vivo 6-h euglycemic hyperinsulinemia; n = 18), and 2) ANGPTL8 and ANGPTL3 gene and protein expression in immortalized human hepatocytes (IHH) and adipocytes were measured. Effect of ANGPTL3 on secretion of ANGPTL8 in cells stably overexpressing ANGPTL3, -8, or both was determined. RESULTS: ANGPTL3 was only expressed in the liver, whereas ANGPTL8 was expressed in both tissues. In vivo hyperinsulinemia significantly decreased both plasma ANGPTL8 and ANGPTL3 at 3 and 6 hours. Insulin increased ANGPTL8 expression in human adipose tissue 14- and 18-fold at 3 and 6 hours and ANGPTL8 was the most insulin-responsive transcript on microarray. Insulin also increased ANPGTL8 in cultured adipocytes and IHH but the protein mainly remained intracellular. In vitro in IHH, insulin decreased ANGPTL3 gene expression and secretion of ANGPTL3 into growth medium. Overexpression of ANGPTL8 in CHO cells did not result in its release into culture medium while abundant secretion occurred in cells co-expressing ANGPTL3 and -8. CONCLUSIONS: Insulin decreases plasma ANGPTL3 by decreasing ANGPTL3 expression in the liver. Insulin markedly increases ANGPTL8 in adipose tissue and the liver but not in plasma. These data show that measurement of plasma ANGPTL3 but not -8 reflects insulin action in target tissues.


Subject(s)
Adipose Tissue/drug effects , Angiopoietins/metabolism , Hepatocytes/drug effects , Insulin/pharmacology , Liver/drug effects , Peptide Hormones/metabolism , Adipose Tissue/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Angiopoietins/genetics , Animals , CHO Cells , Cell Line , Cricetulus , Hepatocytes/metabolism , Humans , Liver/metabolism , Peptide Hormones/genetics
9.
Biosci Rep ; 34(6): e00160, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25495645

ABSTRACT

Homozygosity of loss-of-function mutations in ANGPTL3 (angiopoietin-like protein 3)-gene results in FHBL2 (familial combined hypolipidaemia, OMIM #605019) characterized by the reduction of all major plasma lipoprotein classes, which includes VLDL (very-low-density lipoprotein), LDL (low-density lipoprotein), HDL (high-density lipoprotein) and low circulating NEFAs (non-esterified fatty acids), glucose and insulin levels. Thus complete lack of ANGPTL3 in humans not only affects lipid metabolism, but also affects whole-body insulin and glucose balance. We used wild-type and ANGPTL3-silenced IHHs (human immortalized hepatocytes) to investigate the effect of ANGPTL3 silencing on hepatocyte-specific VLDL secretion and glucose uptake. We demonstrate that both insulin and PPARγ (peroxisome-proliferator-activated receptor γ) agonist rosiglitazone down-regulate the secretion of ANGPTL3 and TAG (triacylglycerol)-enriched VLDL1-type particles in a dose-dependent manner. Silencing of ANGPTL3 improved glucose uptake in hepatocytes by 20-50% and influenced down-regulation of gluconeogenic genes, suggesting that silencing of ANGPTL3 improves insulin sensitivity. We further show that ANGPTL3-silenced cells display a more pronounced shift from the secretion of TAG-enriched VLDL1-type particles to secretion of lipid poor VLDL2-type particles during insulin stimulation. These data suggest liver-specific mechanisms involved in the reported insulin-sensitive phenotype of ANGPTL3-deficient humans, featuring lower plasma insulin and glucose levels.


Subject(s)
Angiopoietin-1/genetics , Gluconeogenesis/genetics , Hepatocytes/drug effects , Insulin/pharmacology , Lipoproteins, VLDL/metabolism , RNA Interference , Triglycerides/metabolism , Angiopoietin-1/metabolism , Cell Line, Transformed , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fatty Acids/metabolism , Fatty Acids/pharmacokinetics , Gene Expression , Glucose/metabolism , Glucose/pharmacokinetics , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Thiazolidinediones/pharmacology
10.
J Clin Endocrinol Metab ; 98(12): 4923-31, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24081738

ABSTRACT

CONTEXT: Obesity is associated with increased circulating 17ß-estradiol (E2), but less is known about E2 concentrations in adipose tissue. In addition to E2, adipose tissue synthesizes E2 fatty acyl esters (E2-FAE). OBJECTIVE: The aim was to compare estrogen concentrations and expression of estrogen-converting enzymes in adipose tissue between severely obese men and women. DESIGN AND SETTING: Tissue samples were obtained during elective surgery in University Central Hospital in the years 2008 through 2011. PATIENTS: We studied 14 men and 22 premenopausal women undergoing bariatric surgery and 10 control women operated for nonmalignant reasons. INTERVENTIONS: Paired samples were taken from abdominal sc and visceral adipose tissue and serum and analyzed for E2 and E2-FAE by fluoroimmunoassay and liquid chromatography-tandem mass spectrometry. mRNA expression of genes was analyzed by quantitative PCR. RESULTS: Compared with men, E2 levels in sc adipose tissue in obese women were higher, along with higher relative mRNA expression of steroid sulfatase and 17ß-hydroxysteroid dehydrogenases 1, 7, and 12. In men, E2-FAE concentrations in adipose tissue were similar to E2 but in women significantly lower compared with E2. Adipose tissue E2-FAE and serum E2-FAE levels correlated positively in obese subjects. Serum E2 did not significantly correlate with E2 concentration or mRNA expression of genes in adipose tissue in obese men or women. CONCLUSIONS: The production of E2 by the large adipose mass was not reflected by increased circulating E2 concentrations in severely obese men or women. However, adipose tissue may contribute to concentrations of serum E2-FAE.


Subject(s)
17-Hydroxysteroid Dehydrogenases/biosynthesis , Estradiol/metabolism , Gene Expression Regulation, Enzymologic , Intra-Abdominal Fat/metabolism , Obesity, Morbid/metabolism , Steryl-Sulfatase/biosynthesis , Subcutaneous Fat, Abdominal/metabolism , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Acylation , Adult , Body Mass Index , Estradiol/analogs & derivatives , Estradiol/blood , Estradiol/chemistry , Fatty Acids/blood , Fatty Acids/chemistry , Fatty Acids/metabolism , Female , Humans , Intra-Abdominal Fat/enzymology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Middle Aged , Obesity, Morbid/blood , Obesity, Morbid/surgery , RNA, Messenger/metabolism , Sex Characteristics , Stereoisomerism , Steryl-Sulfatase/genetics , Steryl-Sulfatase/metabolism , Subcutaneous Fat, Abdominal/enzymology
11.
Transpl Int ; 26(11): 1126-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24102821

ABSTRACT

Heart transplant gene therapy requires vectors with long-lasting gene expression, high cardiotropism, and minimal pathological effects. Here, we examined transduction properties of ex vivo intracoronary delivery of adeno-associated virus (AAV) serotype 2, 8, and 9 in rat syngenic and allogenic heart transplants. Adult Dark Agouti (DA) rat hearts were intracoronarily perfused ex vivo with AAV2, AAV8, or AAV9 encoding firefly luciferase and transplanted heterotopically into the abdomen of syngenic DA or allogenic Wistar-Furth (WF) recipients. Serial in vivo bioluminescent imaging of syngraft and allograft recipients was performed for 6 months and 4 weeks, respectively. Grafts were removed for PCR-, RT-PCR, and luminometer analysis. In vivo bioluminescent imaging of recipients showed that AAV9 induced a prominent and stable luciferase activity in the abdomen, when compared with AAV2 and AAV8. However, ex vivo analyses revealed that intracoronary perfusion with AAV2 resulted in the highest heart transplant transduction levels in syngrafts and allografts. Ex vivo intracoronary delivery of AAV2 resulted in efficient transgene expression in heart transplants, whereas intracoronary AAV9 escapes into adjacent tissues. In terms of cardiac transduction, these results suggest AAV2 as a potential vector for gene therapy in preclinical heart transplants studies, and highlight the importance of delivery route in gene transfer studies.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Heart Transplantation , Heart/virology , Animals , Dependovirus/classification , Isografts , Luciferases/genetics , Male , Myocardium/metabolism , Rats , Transgenes/genetics
12.
Arterioscler Thromb Vasc Biol ; 33(4): 847-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23413431

ABSTRACT

OBJECTIVE: Low high-density lipoprotein cholesterol (HDL-C) is associated with cardiometabolic pathologies. In this study, we investigate the biological pathways and individual genes behind low HDL-C by integrating results from 3 high-throughput data sources: adipose tissue transcriptomics, HDL lipidomics, and dense marker genotypes from Finnish individuals with low or high HDL-C (n=450). APPROACH AND RESULTS: In the pathway analysis of genetic data, we demonstrate that genetic variants within inflammatory pathways were enriched among low HDL-C associated single-nucleotide polymorphisms, and the expression of these pathways upregulated in the adipose tissue of low HDL-C subjects. The lipidomic analysis highlighted the change in HDL particle quality toward putatively more inflammatory and less vasoprotective state in subjects with low HDL-C, as evidenced by their decreased antioxidative plasmalogen contents. We show that the focal point of these inflammatory pathways seems to be the HLA region with its low HDL-associated alleles also associating with more abundant local transcript levels in adipose tissue, increased plasma vascular cell adhesion molecule 1 (VCAM1) levels, and decreased HDL particle plasmalogen contents, markers of adipose tissue inflammation, vascular inflammation, and HDL antioxidative potential, respectively. In a population-based look-up of the inflammatory pathway single-nucleotide polymorphisms in a large Finnish cohorts (n=11 211), no association of the HLA region was detected for HDL-C as quantitative trait, but with extreme HDL-C phenotypes, implying the presence of low or high HDL genes in addition to the population-genomewide association studies-identified HDL genes. CONCLUSIONS: Our study highlights the role of inflammation with a genetic component in subjects with low HDL-C and identifies novel cis-expression quantitative trait loci (cis-eQTL) variants in HLA region to be associated with low HDL-C.


Subject(s)
Adipose Tissue/metabolism , Cholesterol, HDL/blood , Coronary Artery Disease/blood , Coronary Artery Disease/genetics , Gene Expression Profiling , Genomics , Inflammation/blood , Inflammation/genetics , Biomarkers/blood , Coronary Artery Disease/immunology , Female , Finland , Gene Regulatory Networks , Genetic Predisposition to Disease , Genome-Wide Association Study , HLA Antigens/genetics , Health Surveys , Humans , Inflammation/immunology , Linear Models , Logistic Models , Male , Middle Aged , Phenotype , Plasmalogens/blood , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Risk Factors , Vascular Cell Adhesion Molecule-1/blood
13.
Dis Model Mech ; 6(2): 342-57, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23065637

ABSTRACT

Both CLN1 and CLN5 deficiencies lead to severe neurodegenerative diseases of childhood, known as neuronal ceroid lipofuscinoses (NCLs). The broadly similar phenotypes of NCL mouse models, and the potential for interactions between NCL proteins, raise the possibility of shared or convergent disease mechanisms. To begin addressing these issues, we have developed a new mouse model lacking both Cln1 and Cln5 genes. These double-knockout (Cln1/5 dko) mice were fertile, showing a slight decrease in expected Mendelian breeding ratios, as well as impaired embryoid body formation by induced pluripotent stem cells derived from Cln1/5 dko fibroblasts. Typical disease manifestations of the NCLs, i.e. seizures and motor dysfunction, were detected at the age of 3 months, earlier than in either single knockout mouse. Pathological analyses revealed a similar exacerbation and earlier onset of disease in Cln1/5 dko mice, which exhibited a pronounced accumulation of autofluorescent storage material. Cortical demyelination and more pronounced glial activation in cortical and thalamic regions was followed by cortical neuron loss. Alterations in lipid metabolism in Cln1/5 dko showed a specific increase in plasma phospholipid transfer protein (PLTP) activity. Finally, gene expression profiling of Cln1/5 dko cortex revealed defects in myelination and immune response pathways, with a prominent downregulation of α-synuclein in Cln1/5 dko mouse brains. The simultaneous loss of both Cln1 and Cln5 genes might enhance the typical pathological phenotypes of these mice by disrupting or downregulating shared or convergent pathogenic pathways, which could potentially include interactions of CLN1 and CLN5.


Subject(s)
Membrane Glycoproteins/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Thiolester Hydrolases/metabolism , Aging/pathology , Animals , Cell Differentiation , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebral Cortex/ultrastructure , Embryoid Bodies/metabolism , Embryoid Bodies/pathology , Female , Fluorescence , Gene Expression Profiling , Gliosis , Immunity/immunology , Induced Pluripotent Stem Cells/metabolism , Lysosomal Membrane Proteins , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout , Myelin Sheath/metabolism , Myelin Sheath/pathology , Neuronal Ceroid-Lipofuscinoses/blood , Neuronal Ceroid-Lipofuscinoses/immunology , Neurons/metabolism , Neurons/pathology , Phenotype , Phospholipid Transfer Proteins/blood , Phospholipids/blood , Thiolester Hydrolases/deficiency , alpha-Synuclein/metabolism
14.
BMC Med Genomics ; 5: 9, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22471940

ABSTRACT

BACKGROUND: To get insight into molecular mechanisms underlying insulin resistance, we compared acute in vivo effects of insulin on adipose tissue transcriptional profiles between obese insulin-resistant and lean insulin-sensitive women. METHODS: Subcutaneous adipose tissue biopsies were obtained before and after 3 and 6 hours of intravenously maintained euglycemic hyperinsulinemia from 9 insulin-resistant and 11 insulin-sensitive females. Gene expression was measured using Affymetrix HG U133 Plus 2 microarrays and qRT-PCR. Microarray data and pathway analyses were performed with Chipster v1.4.2 and by using in-house developed nonparametric pathway analysis software. RESULTS: The most prominent difference in gene expression of the insulin-resistant group during hyperinsulinemia was reduced transcription of nuclear genes involved in mitochondrial respiration (mitochondrial respiratory chain, GO:0001934). Inflammatory pathways with complement components (inflammatory response, GO:0006954) and cytokines (chemotaxis, GO:0042330) were strongly up-regulated in insulin-resistant as compared to insulin-sensitive subjects both before and during hyperinsulinemia. Furthermore, differences were observed in genes contributing to fatty acid, cholesterol and triglyceride metabolism (FATP2, ELOVL6, PNPLA3, SREBF1) and in genes involved in regulating lipolysis (ANGPTL4) between the insulin-resistant and -sensitive subjects especially during hyperinsulinemia. CONCLUSIONS: The major finding of this study was lower expression of mitochondrial respiratory pathway and defective induction of lipid metabolism pathways by insulin in insulin-resistant subjects. Moreover, the study reveals several novel genes whose aberrant regulation is associated with the obese insulin-resistant phenotype.


Subject(s)
Insulin Resistance/genetics , Insulin/metabolism , Mitochondria/metabolism , Obesity/genetics , Subcutaneous Fat/metabolism , Adolescent , Adult , Female , Gene Expression , Humans , Inflammation/genetics , Inflammation/metabolism , Lipid Metabolism/genetics , Middle Aged , Mitochondria/genetics , Obesity/metabolism , RNA, Messenger/metabolism
15.
Circulation ; 122(17): 1725-33, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20937974

ABSTRACT

BACKGROUND: Vascular endothelial growth factor-B (VEGF-B) binds to VEGF receptor-1 and neuropilin-1 and is abundantly expressed in the heart, skeletal muscle, and brown fat. The biological function of VEGF-B is incompletely understood. METHODS AND RESULTS: Unlike placenta growth factor, which binds to the same receptors, adeno-associated viral delivery of VEGF-B to mouse skeletal or heart muscle induced very little angiogenesis, vascular permeability, or inflammation. As previously reported for the VEGF-B(167) isoform, transgenic mice and rats expressing both isoforms of VEGF-B in the myocardium developed cardiac hypertrophy yet maintained systolic function. Deletion of the VEGF receptor-1 tyrosine kinase domain or the arterial endothelial Bmx tyrosine kinase inhibited hypertrophy, whereas loss of VEGF-B interaction with neuropilin-1 had no effect. Surprisingly, in rats, the heart-specific VEGF-B transgene induced impressive growth of the epicardial coronary vessels and their branches, with large arteries also seen deep inside the subendocardial myocardium. However, VEGF-B, unlike other VEGF family members, did not induce significant capillary angiogenesis, increased permeability, or inflammatory cell recruitment. CONCLUSIONS: VEGF-B appears to be a coronary growth factor in rats but not in mice. The signals for the VEGF-B-induced cardiac hypertrophy are mediated at least in part via the endothelium. Because cardiomyocyte damage in myocardial ischemia begins in the subendocardial myocardium, the VEGF-B-induced increased arterial supply to this area could have therapeutic potential in ischemic heart disease.


Subject(s)
Capillary Permeability/physiology , Coronary Vessels/growth & development , Inflammation/physiopathology , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor B/physiology , Adenoviridae/genetics , Animals , Cardiomegaly/physiopathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Models, Animal , Muscle, Skeletal/blood supply , Myocardium , Neuropilin-1/physiology , Rats , Rats, Transgenic , Rats, Wistar , Vascular Endothelial Growth Factor B/genetics
16.
Circ Res ; 104(11): 1302-12, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19443835

ABSTRACT

The therapeutic potential of vascular endothelial growth factor (VEGF)-C and VEGF-D in skeletal muscle has been of considerable interest as these factors have both angiogenic and lymphangiogenic activities. Previous studies have mainly used adenoviral gene delivery for short-term expression of VEGF-C and VEGF-D in pig, rabbit, and mouse skeletal muscles. Here we have used the activated mature forms of VEGF-C and VEGF-D expressed via recombinant adeno-associated virus (rAAV), which provides stable, long-lasting transgene expression in various tissues including skeletal muscle. Mouse tibialis anterior muscle was transduced with rAAV encoding human or mouse VEGF-C or VEGF-D. Two weeks later, immunohistochemical analysis showed increased numbers of both blood and lymph vessels, and Doppler ultrasound analysis indicated increased blood vessel perfusion. The lymphatic vessels further increased at the 4-week time point were functional, as shown by FITC-lectin uptake and transport. Furthermore, receptor activation and arteriogenic activity were increased by an alanine substitution mutant of human VEGF-C (C137A) having an increased dimer stability and by a chimeric CAC growth factor that contained the VEGF receptor-binding domain flanked by VEGF-C propeptides, but only the latter promoted significantly more blood vessel perfusion when compared to the other growth factors studied. We conclude that long-term expression of VEGF-C and VEGF-D in skeletal muscle results in the generation of new functional blood and lymphatic vessels. The therapeutic value of intramuscular lymph vessels in draining tissue edema and lymphedema can now be evaluated using this model system.


Subject(s)
Blood Vessels/physiology , Heart/physiology , Lymphatic Vessels/physiology , Muscle, Skeletal/physiology , Vascular Endothelial Growth Factor B/physiology , Vascular Endothelial Growth Factor C/physiology , Animals , Dimerization , Drug Stability , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/blood supply , Mutation , Polymorphism, Single Nucleotide , Recombinant Proteins/metabolism , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-2/physiology
17.
Arterioscler Thromb Vasc Biol ; 29(5): 691-8, 2009 May.
Article in English | MEDLINE | ID: mdl-19213942

ABSTRACT

OBJECTIVE: Chronic rejection is the main reason for the poor long-term survival of heart transplant recipients and is characterized by cardiac allograft inflammation, fibrosis, and arteriosclerosis. We examined the specific roles of different platelet-derived growth factor (PDGF) ligands (A-D)--potent mesenchymal cell mitogens--in rat cardiac allografts. METHODS AND RESULTS: PDGFR-alpha mRNA was upregulated in acutely-rejecting, and PDGF-A and PDGF-C mRNA in chronically-rejecting cardiac centhatn allografts. In acute rejection, PDGFR-alpha immunoreactivity increased in the media of arteries. In chronically-rejecting allografts, immunoreactivity of all PDGF ligands and receptors--except that of PDGF-B ligand--was found in the intima of arteries, and the expression of PDGF-A and PDGF-C was seen in cardiomyocytes. Intracoronary adeno-associated virus-2 (AAV2)-mediated PDGF-A and -D gene transfer enhanced cardiac allograft inflammation. AAV2-PDGF-A, AAV2-PDGF-C, and AAV2-PDGF-D significantly upregulated profibrotic TGF-beta1 mRNA and accelerated cardiac fibrosis and arteriosclerosis. In contrast, AAV2-PDGF-B did not aggravate chronic rejection. CONCLUSIONS: We found that alloimmune response induces PDGF-A, PDGF-C, and PDGF-D expression in the graft vasculature. PDGF-A, PDGF-C, and PDGF-D mediated profibrotic and proarteriosclerotic effects in transplanted hearts involving the TGF-beta1 pathway. Inhibition of signaling of all PDGF-ligands except that of PDGF-B may thus be needed to inhibit chronic rejection in cardiac allografts.


Subject(s)
Arteriosclerosis/immunology , Graft Rejection/metabolism , Heart Transplantation/immunology , Platelet-Derived Growth Factor/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Disease Models, Animal , Fibrosis/immunology , Graft Rejection/immunology , Graft Rejection/pathology , RNA, Messenger/metabolism , Rats , Signal Transduction , Transplantation, Homologous , Up-Regulation
18.
FASEB J ; 19(10): 1365-7, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15958522

ABSTRACT

Therapeutic angiogenesis provides a potential alternative for the treatment of cardiovascular ischemic diseases. Vascular endothelial growth factor (VEGF) is an important component of the angiogenic response to ischemia. Here we used adeno-associated virus (AAV) gene delivery to skeletal muscle to examine the effects of VEGF vs. a stabilized form of hypoxia-inducible factor-1alpha (HIF-1alpha). The recombinant AAVs were injected into mouse tibialis anterior muscle, and their effects were analyzed by immunohistochemistry and functional assays. These analyses showed that stabilized HIF-1alpha markedly increase capillary sprouting and proliferation, whereas VEGF164 or VEGF120 induced only proliferation of endothelial cells without formation of proper capillary structures. The Evans Blue permeability assay indicated that, unlike VEGF, HIF-1alpha overexpression did not increase vascular leakiness in the transduced muscle. Doppler ultrasound imaging showed that vascular perfusion in the HIF-1alpha treated muscles was significantly enhanced when compared to the controls and not further improved by co-expression of the arteriogenic growth factors angiopoietin-1 or platelet-derived growth factor-B. Our results show that AAV-mediated transduction of a stabilized form of HIF-1alpha can circumvent the problems associated with overexpression of individual angiogenic growth factors. HIF-1alpha should thus offer a potent alternative for pro-angiogenic gene therapy.


Subject(s)
Dependovirus/genetics , Gene Transfer, Horizontal , Genetic Therapy , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/genetics , Animals , Capillary Permeability , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Perfusion , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Proto-Oncogene Proteins c-sis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...