Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
PLoS Pathog ; 10(12): e1004534, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25502789

ABSTRACT

Apolipoprotein B (ApoB) and ApoE have been shown to participate in the particle formation and the tissue tropism of hepatitis C virus (HCV), but their precise roles remain uncertain. Here we show that amphipathic α-helices in the apolipoproteins participate in the HCV particle formation by using zinc finger nucleases-mediated apolipoprotein B (ApoB) and/or ApoE gene knockout Huh7 cells. Although Huh7 cells deficient in either ApoB or ApoE gene exhibited slight reduction of particles formation, knockout of both ApoB and ApoE genes in Huh7 (DKO) cells severely impaired the formation of infectious HCV particles, suggesting that ApoB and ApoE have redundant roles in the formation of infectious HCV particles. cDNA microarray analyses revealed that ApoB and ApoE are dominantly expressed in Huh7 cells, in contrast to the high level expression of all of the exchangeable apolipoproteins, including ApoA1, ApoA2, ApoC1, ApoC2 and ApoC3 in human liver tissues. The exogenous expression of not only ApoE, but also other exchangeable apolipoproteins rescued the infectious particle formation of HCV in DKO cells. In addition, expression of these apolipoproteins facilitated the formation of infectious particles of genotype 1b and 3a chimeric viruses. Furthermore, expression of amphipathic α-helices in the exchangeable apolipoproteins facilitated the particle formation in DKO cells through an interaction with viral particles. These results suggest that amphipathic α-helices in the exchangeable apolipoproteins play crucial roles in the infectious particle formation of HCV and provide clues to the understanding of life cycle of HCV and the development of novel anti-HCV therapeutics targeting for viral assembly.


Subject(s)
Apolipoproteins B/chemistry , Apolipoproteins B/physiology , Apolipoproteins E/chemistry , Apolipoproteins E/physiology , Hepacivirus/pathogenicity , Protein Structure, Secondary/physiology , Virion/pathogenicity , Apolipoproteins A/physiology , Apolipoproteins B/genetics , Apolipoproteins C/physiology , Apolipoproteins E/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Gene Expression Regulation, Viral/drug effects , Gene Knockout Techniques , Hepacivirus/physiology , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , RNA, Small Interfering/pharmacology , Virion/physiology , Virus Replication/physiology
2.
Circulation ; 114(7): 681-7, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16894036

ABSTRACT

BACKGROUND: Activation of vascular endothelial cells (ECs) plays an important role in atherogenesis and plaque instability. Lipoproteins containing apolipoprotein CIII (apoCIII) predict coronary heart disease (CHD). We recently reported that apoCIII has a proinflammatory effect on human monocytes. In this study, we looked for a direct effect of apoCIII on EC expression of adhesion molecules, leading to monocytic cell adhesion. METHODS AND RESULTS: Treatment of ECs with apoCIII or apoCIII-rich VLDL caused human monocytic THP-1 cells to adhere to them under static condition or under laminar sheer stress (1.0 dyne/cm2). ApoCIII increased EC expression of vascular cell adhesion molecule-1 (VCAM-1) protein and intercellular cell adhesion molecule-1 (ICAM-1) protein (4.9 +/- 1.5-fold and 1.4 +/- 0.5-fold versus control, respectively). Furthermore, apoCIII remarkably increased membrane-bound protein kinase C (PKC) beta in ECs, indicating activation. A selective inhibitor of PKCbeta prevented the rise in VCAM-1 and THP-1 cell adhesion to ECs. Moreover, exposure of ECs to apoCIII induced nuclear factor-kappaB (NF-kappaB) activation. PKCbeta inhibition abolished apoCIII-induced NF-kappaB activation, and NF-kappaB inhibition reduced expression of VCAM-1, each resulting in reduced THP-1 cell adhesion. ApoCIII-rich VLDL also activated PKCbeta and NF-kappaB in ECs and increased expression of VCAM-1. Pretreatment of ApoCIII-rich VLDL with anti-apoCIII neutralizing antibody abolished its effect on PKCbeta activation. CONCLUSIONS: Our findings provide the first evidence that apoCIII increases VCAM-1 and ICAM-1 expression in ECs by activating PKCbeta and NF-kappaB, suggesting a novel mechanism for EC activation induced by dyslipidemia. Therefore, apoCIII-rich VLDL may contribute directly to atherogenesis by activating ECs and recruiting monocytes to them.


Subject(s)
Apolipoproteins C/physiology , Endothelial Cells/metabolism , Monocytes/drug effects , Vascular Cell Adhesion Molecule-1/metabolism , Apolipoprotein C-III , Atherosclerosis/physiopathology , Cell Adhesion/drug effects , Cell Line , Cells, Cultured , Cholesterol, VLDL/blood , Cholesterol, VLDL/pharmacology , Dyslipidemias/complications , Endothelial Cells/cytology , Humans , I-kappa B Proteins/metabolism , Integrin beta1/physiology , Intercellular Adhesion Molecule-1/metabolism , Monocytes/cytology , NF-KappaB Inhibitor alpha , NF-kappa B/physiology , Protein Kinase C/physiology
3.
J Lipid Res ; 47(6): 1203-11, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16537968

ABSTRACT

Previous studies have shown that overexpression of human apolipoprotein C-I (apoC-I) results in moderate hypercholesterolemia and severe hypertriglyceridemia in mice in the presence and absence of apoE. We assessed whether physiological endogenous apoC-I levels are sufficient to modulate plasma lipid levels independently of effects of apoE on lipid metabolism by comparing apolipoprotein E gene-deficient/apolipoprotein C-I gene-deficient (apoe-/-apoc1-/-), apoe-/-apoc1+/-, and apoe-/-apoc1+/+ mice. The presence of the apoC-I gene-dose-dependently increased plasma cholesterol (+45%; P < 0.001) and triglycerides (TGs) (+137%; P < 0.001), both specific for VLDL. Whereas apoC-I did not affect intestinal [3H]TG absorption, it increased the production rate of hepatic VLDL-TG (+35%; P < 0.05) and VLDL-[35S]apoB (+39%; P < 0.01). In addition, apoC-I increased the postprandial TG response to an intragastric olive oil load (+120%; P < 0.05) and decreased the uptake of [3H]TG-derived FFAs from intravenously administered VLDL-like emulsion particles by gonadal and perirenal white adipose tissue (WAT) (-34% and -25%, respectively; P < 0.05). As LPL is the main enzyme involved in the clearance of TG-derived FFAs by WAT, and total postheparin plasma LPL levels were unaffected, these data demonstrate that endogenous apoC-I suffices to attenuate the lipolytic activity of LPL. Thus, we conclude that endogenous plasma apoC-I increases VLDL-total cholesterol and VLDL-TG dose-dependently in apoe-/- mice, resulting from increased VLDL particle production and LPL inhibition.


Subject(s)
Apolipoproteins C/physiology , Apolipoproteins E/physiology , Hyperlipidemias/metabolism , Lipoprotein Lipase/metabolism , Lipoproteins, VLDL/biosynthesis , Animals , Apolipoprotein C-I , Apolipoproteins C/blood , Apolipoproteins C/genetics , Apolipoproteins E/blood , Apolipoproteins E/genetics , Cholesterol/blood , Cholesterol/metabolism , Hyperlipidemias/blood , Intestinal Mucosa/metabolism , Lipase/metabolism , Lipoprotein Lipase/analysis , Lipoprotein Lipase/blood , Lipoproteins, VLDL/analysis , Lipoproteins, VLDL/blood , Liver/metabolism , Mice , Mice, Knockout , Postprandial Period
4.
J Lipid Res ; 47(6): 1212-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16495512

ABSTRACT

Apolipoprotein C-III (apoC-III) production rate (PR) is strongly correlated with plasma triglyceride (TG) levels. ApoC-III exists in three different isoforms, according to the sialylation degree of the protein. We investigated the kinetics and respective role of each apoC-III isoform in modulating intravascular lipid/lipoprotein metabolism. ApoC-III kinetics were measured in a sample of 18 healthy men [mean age (+/-SD) 42.1 +/- 9.5 years, body mass index 29.8 +/- 4.6 kg/m2] using a primed-constant infusion of l-(5,5,5-D3) leucine for 12 h. Mono-sialylated and di-sialylated apoC-III (apo-CIII1 and apoC-III2) exhibited similar PRs (means +/- SD, 1.22 +/- 0.49 mg/kg/day vs. 1.15 +/- 0.59 mg/kg/day, respectively) and similar fractional catabolic rates (FCRs) (0.51 +/- 0.13 pool/day vs. 0.61 +/- 0.24 pool/day, respectively). Nonsialylated apoC-III (apoC-III0) had an 80% lower PR (0.25 +/- 0.12 mg/kg/day) and a 60% lower FCR (0.21 +/- 0.07 pool/day) (P < 0.0001 for comparison with CIII1 and CIII2 isoforms). The PRs of apoC-III1 and apoC-III2 were more strongly correlated with plasma TG levels (r > 0.8, P < 0.0001) than was apoC-III0 PR (r = 0.54, P < 0.05). Finally, the PR of apoC-III2 was strongly correlated with the proportion of LDL <255 A (r = 0.72, P = 0.002). These results suggest that all apoC-III isoforms, especially the predominant CIII1 and CIII2 isoforms, contribute to hypertriglyceridemia and that apoC-III2 may play a significant role in the expression of the small, dense LDL phenotype.


Subject(s)
Apolipoproteins C/metabolism , Lipid Metabolism/physiology , Adult , Apolipoprotein C-III , Apolipoproteins C/blood , Apolipoproteins C/physiology , Cholesterol/blood , Cholesterol/metabolism , Humans , Kinetics , Lipoproteins, HDL/blood , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/blood , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/metabolism , Male , Middle Aged , Protein Isoforms/blood , Protein Isoforms/metabolism , Protein Isoforms/physiology , Statistics as Topic
5.
Circulation ; 113(5): 691-700, 2006 Feb 07.
Article in English | MEDLINE | ID: mdl-16461842

ABSTRACT

BACKGROUND: Lipoproteins containing apolipoprotein (apo) CIII predict coronary heart disease and associate with components of the metabolic syndrome. ApoCIII inhibits lipoprotein catabolism in plasma. However, it is unknown whether apoCIII itself, or in association with VLDL, LDL, or HDL, directly affects atherogenic mechanisms in vascular cells. Thus, we investigated the direct effect of lipoproteins that do or do not have apoCIII, and apoCIII itself, on adhesion of THP-1 cells, a human monocytic cell line, to vascular endothelial cells (ECs). METHODS AND RESULTS: VLDL CIII+ and LDL CIII+ (100 microg apoB/mL) from fasting plasma of 18 normolipidemic volunteers increased THP-1 cell adhesion to ECs under static conditions by 2.4+/-0.3-fold and 1.8+/-0.7-fold, respectively (P<0.01), whereas VLDL or LDL without apoCIII did not affect THP-1 cell adhesion. ApoCIII (100 microg/mL), but not apoCI, apoCII or apoE, also increased THP-1 cell adhesion by 2.1+/-0.6-fold. Studies with human peripheral blood monocytes yielded similar results. ApoCIII also had strong proadhesive effects under shear flow conditions. VLDL CIII+, LDL CIII+, or apoCIII itself activated PKCalpha and RhoA in THP-1 cells, which resulted in beta1-integrin activation and enhancement of THP-1 cell adhesion. Interestingly, HDL CIII+ did not affect THP-1 cell adhesion, whereas HDL without apoCIII decreased their adhesion. CONCLUSIONS: ApoB lipoproteins that contain apoCIII increase THP-1 cell adhesion to ECs via PKCalpha and RhoA-mediated beta1-integrin activation. These results indicate that apoCIII not only modulates lipoprotein metabolism but also may directly contribute to the development of atherosclerosis.


Subject(s)
Apolipoproteins B , Apolipoproteins C/physiology , Endothelium, Vascular/cytology , Lipoproteins/chemistry , Monocytes/cytology , Apolipoprotein C-III , Atherosclerosis/etiology , Cell Adhesion , Humans , Integrin beta1/metabolism , Lipoproteins, LDL/blood , Lipoproteins, LDL/chemistry , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/chemistry , Protein Kinase C-alpha , rhoA GTP-Binding Protein
7.
Acta Physiol Hung ; 92(2): 109-20, 2005.
Article in English | MEDLINE | ID: mdl-16268049

ABSTRACT

Both epidemiological and intervention studies have shown that hypertriglyceridemia is a significant cardiovascular risk factor. The large variation of the triglyceride values is explained by the influence of several modifying factors, which are difficult to standardise. Therefore hypertriglyceridemia should be considered rather as risk marker, than risk factor. The measurement of the apolipoprotein CIII level, which is a more stable parameter of the triglyceride rich lipid particles, is now becoming more widespread. This parameter is also able to substitute the assessment of the small dense LDL form that has a controversial significance. The clinical benefit of reduction of triglyceride concentration and the accompanying increase of HDL cholesterol level by fibrates, in the prevention of the coronary heart disease (CHD) events, have been demonstrated in several prospective, placebo-controlled trials. The VA-HIT study, enrolling the largest number of patients, has shown that fibrates have another effect, presumably influencing the insulin resistance independently of lipid levels that is also able to reduce the CHD events.


Subject(s)
Coronary Disease/etiology , Hypertriglyceridemia/complications , Apolipoprotein C-III , Apolipoproteins C/physiology , Clinical Trials as Topic , Clofibric Acid/pharmacology , Clofibric Acid/therapeutic use , Humans , Hyperinsulinism/drug therapy , Hypertriglyceridemia/drug therapy , Insulin Resistance , Lipoproteins/metabolism , Lipoproteins, HDL/physiology , Lipoproteins, LDL/physiology , Obesity/complications , Risk , Triglycerides/blood
8.
Int J Biochem Cell Biol ; 37(6): 1308-18, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15778093

ABSTRACT

Plasma low- and high-density lipoproteins (LDL and HDL) are cleared from the circulation by specific receptors and are either totally degraded or their cholesteryl esters (CE) are selectively delivered to cells by receptors such as the scavenger receptor class B type I (SR-BI). The aim of the present study was to define the effect of apoC-II and apoC-III on the uptake of LDL and HDL by HepG2 cells. Stable transformants were obtained with sense or antisense strategies that secrete 47-294% the normal level of apoC-II or 60-200% that of apoC-III. Different levels of secreted apoC-II or apoC-III had little effect on LDL and HDL protein degradation by HepG2 cells. However, compared to controls, cells under-expressing apoC-II showed a 160% higher capacity to selectively take up HDL-CE, while cells under-expressing apoC-III demonstrated 70 and 160% higher capacity to take up CE from LDL and HDL, respectively. In experiments conducted with exogenously added apoC-II or apoC-III, no significant effect was observed on lipoprotein-protein association/degradation; however, LDL-CE and HDL-CE selective uptake was significantly reduced in a dose-dependent manner. These results indicate that apoC-II and apoC-III inhibit CE-selective uptake.


Subject(s)
Apolipoproteins C/physiology , Cholesterol, HDL/antagonists & inhibitors , Cholesterol, HDL/metabolism , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/metabolism , Apolipoprotein C-II , Apolipoprotein C-III , Apolipoproteins C/metabolism , CD36 Antigens , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Humans , Receptors, Immunologic/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B
9.
Diabetes ; 54(3): 664-71, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15734841

ABSTRACT

Our aim was to study whether the absence of apolipoprotein (apo) C3, a strong inhibitor of lipoprotein lipase (LPL), accelerates the development of obesity and consequently insulin resistance. Apoc3(-/-) mice and wild-type littermates were fed a high-fat (46 energy %) diet for 20 weeks. After 20 weeks of high-fat feeding, apoc3(-/-) mice showed decreased plasma triglyceride levels (0.11 +/- 0.02 vs. 0.29 +/- 0.04 mmol, P < 0.05) and were more obese (42.8 +/- 3.2 vs. 35.2 +/- 3.3 g; P < 0.05) compared with wild-type littermates. This increase in body weight was entirely explained by increased body lipid mass (16.2 +/- 5.9 vs. 10.0 +/- 1.8 g; P < 0.05). LPL-dependent uptake of triglyceride-derived fatty acids by adipose tissue was significantly higher in apoc3(-/-) mice. LPL-independent uptake of albumin-bound fatty acids did not differ. It is interesting that whole-body insulin sensitivity using hyperinsulinemic-euglycemic clamps was decreased by 43% and that suppression of endogenous glucose production was decreased by 25% in apoc3(-/-) mice compared with control mice. Absence of apoC3, the natural LPL inhibitor, enhances fatty acid uptake from plasma triglycerides in adipose tissue, which leads to higher susceptibility to diet-induced obesity followed by more severe development of insulin resistance. Therefore, apoC3 is a potential target for treatment of obesity and insulin resistance.


Subject(s)
Apolipoproteins C/physiology , Insulin Resistance/physiology , Obesity/physiopathology , Adipose Tissue/metabolism , Animals , Apolipoprotein C-III , Apolipoproteins C/deficiency , Apolipoproteins C/genetics , Blood Glucose/metabolism , Dietary Fats , Fatty Acids/metabolism , Female , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Time Factors , Triglycerides/blood
10.
Proc Natl Acad Sci U S A ; 101(27): 10090-4, 2004 Jul 06.
Article in English | MEDLINE | ID: mdl-15210953

ABSTRACT

In type 1 diabetes (T1D), there is a specific destruction of the insulin secreting pancreatic beta cell. Although the exact molecular mechanisms underlying beta cell destruction are not known, sera from T1D patients have been shown to promote Ca(2+)-induced apoptosis. We now demonstrate that apolipoprotein CIII (apoCIII) is increased in serum from T1D patients and that this serum factor both induces increased cytoplasmic free intracellular Ca(2+) concentration ([Ca(2+)](i)) and beta cell death. The apoCIII-induced increase in [Ca(2+)](i) reflects an activation of the voltage-gated L-type Ca(2+) channel. Both the effects of T1D sera and apoCIII on the beta cell are abolished in the presence of antibody against apoCIII. Increased serum levels of apoCIII can thus account for the increase in beta cell [Ca(2+)](i) and thereby beta cell apoptosis associated with T1D.


Subject(s)
Apolipoproteins C/physiology , Apoptosis , Calcium/physiology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Adult , Animals , Apolipoprotein C-III , Calcium Channels, L-Type/physiology , Diabetes Mellitus, Type 1/blood , Female , Humans , Male , Mice
11.
Arterioscler Thromb Vasc Biol ; 24(7): 1297-302, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15117734

ABSTRACT

OBJECTIVE: Both the apolipoprotein A5 and C3 genes have repeatedly been shown to play an important role in determining plasma triglyceride concentrations in humans and mice. In mice, transgenic and knockout experiments indicate that plasma triglyceride levels are strongly altered by changes in the expression of either of these 2 genes. In humans, common polymorphisms in both genes have also been associated with plasma triglyceride concentrations. These similar findings raised the issue of the relationship between these 2 genes and altered triglycerides. METHODS AND RESULTS: To address this issue, we generated independent lines of mice that either overexpressed ("double transgenic") or completely lacked ("double knockout") both apolipoprotein genes. We report that both "double transgenic" and "double knockout" mice display normal triglyceride concentrations compared with overexpression or deletion of either gene alone. Furthermore, we find that human ApoAV plasma protein levels in the "double transgenic" mice are approximately 500-fold lower than human ApoCIII levels, supporting ApoAV as a potent triglyceride modulator despite its low concentration. CONCLUSIONS: Together, these data support that APOA5 and APOC3 independently influence plasma triglyceride concentrations but in an opposing manner.


Subject(s)
Apolipoproteins C/blood , Apolipoproteins/blood , Triglycerides/blood , Animals , Apolipoprotein A-V , Apolipoprotein C-III , Apolipoproteins/genetics , Apolipoproteins/physiology , Apolipoproteins A , Apolipoproteins C/genetics , Apolipoproteins C/physiology , Female , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/physiology
12.
Neurogenetics ; 4(3): 141-6, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12736801

ABSTRACT

Previous research has shown that polymorphisms of the apolipoproteins E ( APOE) and APOC1 represent genetic risk factors for dementia and for cognitive impairment in the elderly. The brain mechanisms by which these genetic variations affect behavior or clinical severity are poorly understood. We studied the effect of APOE and APOC1 genes on magnetic resonance imaging measures in a sample of 50 subjects with age-associated memory impairment. The APOE E4 allele was associated with reduced left hippocampal volumes and APOE*E3 status was associated with greater frontal lobe white matter volumes. However, no APOE effects were observed when analyses accounted for other potential confounding variables. The effects of APOC1 on hippocampal volumes appeared to be more robust than those of the APOE polymorphism. However, no modulatory effects on brain morphology outside the medial temporal lobe region were observed when demographic variables, clinical status, and other anatomical brain measurements were taken into consideration. Our results suggest that the role of the APOC1 polymorphism in brain morphology of the cognitively impaired elderly should be examined in further studies.


Subject(s)
Aging/psychology , Apolipoproteins C/genetics , Apolipoproteins E/genetics , Brain/pathology , Memory Disorders/pathology , Polymorphism, Genetic , Aged , Aging/pathology , Alleles , Apolipoprotein C-I , Apolipoprotein E2 , Apolipoprotein E3 , Apolipoprotein E4 , Apolipoproteins C/physiology , Apolipoproteins E/physiology , Cephalometry , Cerebral Ventricles/pathology , Confounding Factors, Epidemiologic , Female , Frontal Lobe/pathology , Genetic Predisposition to Disease , Genotype , Hippocampus/pathology , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Temporal Lobe/pathology , Verbal Learning
13.
J Lipid Res ; 43(3): 416-27, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11893778

ABSTRACT

We conducted a cross-sectional study in a Spanish population (n = 1,029) to investigate associations between the LPL and APOC3 gene loci (LPL-HindIII, LPL-S447X, and APOC3-SstI) and plasma lipid levels and their interaction with APOE polymorphisms and smoking. Carriers of the H(-) or the X447 allele had higher levels of HDL cholesterol (HDL-C), and lower levels of TG, after adjustment for age, body mass index, alcohol, smoking, exercise, and education (P < 0.01). The APOC3 polymorphism presented additive effects to the LPL variants on TG and HDL-C levels in men, and on TG in women. The most and the least favorable haplotype combinations were H(-)/X447/S1 and H(+)/S447/S2, respectively. These combinations accounted for 7% and 5% of the variation in HDL-C and TG in men, and 3% and 4% in women. There was a significant interaction between APOE and LPL variants and HDL-C levels in both genders (P < 0.05). The increases in HDL-C observed for the rare alleles were higher in epsilon4 than in epsilon3 subjects, and absent in epsilon2 individuals. This effect was modulated by smoking (interaction HindIII-APOE-smoking, P = 0.019), indicating that smoking abolished the increase in HDL-C levels observed in epsilon4/H(-) subjects. Understanding this gene-gene-environmental interaction may facilitate preventive interventions to reduce coronary artery disease risk.


Subject(s)
Apolipoproteins C/genetics , Apolipoproteins E/genetics , Lipids/blood , Lipoprotein Lipase/genetics , Polymorphism, Genetic/genetics , Smoking/genetics , Adult , Apolipoproteins C/physiology , Cholesterol, HDL/blood , Cross-Sectional Studies , Deoxyribonuclease HindIII/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , Female , Genetic Variation/genetics , Humans , Lipoprotein Lipase/physiology , Male , Polymorphism, Genetic/physiology , Spain/epidemiology , Triglycerides/blood
14.
Horm Metab Res ; 34(1): 21-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11832997

ABSTRACT

In this study, we examined glucose homeostasis and insulin secretion in transgenic mice overexpressing the human apolipoprotein CIII gene (apo CIII tg). These mice have elevated plasma levels of triglycerides, FFA and cholesterol compared to control mice. The body weight, plasma glucose, and insulin levels, glucose disappearance rates, areas under the ipGTT curve for adult (4 - 8 mo. old) and aged (20 - 24 mo. old) apo CIII tg mice and the determination of insulin during the ipGTT were not different from those of control mice. However, an additional elevation of plasma FFA by treatment with heparin for 2 - 4 h impaired the ipGTT responses in apo CIII tg mice compared to saline-treated mice. The glucose disappearance rate in heparin-treated transgenic mice was slightly lower than in heparin-treated controls. Glucose (22.2 mmol/l) stimulated insulin secretion in isolated islets to the same extent in saline-treated control and apo CIII tg mice. In islets from heparin-treated apo CIII tg mice, the insulin secretion at 2.8 and 22.2 mmol glucose/l was lower than in heparin-treated control mice. In conclusion, hypertriglyceridemia per se or a mild elevation in FFA did not affect insulin secretion or insulin resistance in adult or aged apo CIII tg mice. Nonetheless, an additional elevation of FFA induced by heparin in hypertriglyceridemic mice impaired the ipGTT by reducing insulin secretion.


Subject(s)
Blood Glucose/metabolism , Hypertriglyceridemia/blood , Insulin/metabolism , Islets of Langerhans/metabolism , Animals , Apolipoprotein C-III , Apolipoproteins C/physiology , Area Under Curve , Body Weight , Cholesterol/blood , Fatty Acids, Nonesterified , Female , Glucose Tolerance Test , Heparin/administration & dosage , Hypertriglyceridemia/physiopathology , Insulin/blood , Insulin/physiology , Insulin Secretion , Male , Mice , Mice, Transgenic , Statistics, Nonparametric , Triglycerides/blood
15.
J Biol Chem ; 277(6): 4334-42, 2002 Feb 08.
Article in English | MEDLINE | ID: mdl-11719505

ABSTRACT

Apolipoprotein CII (apoCII) activates lipoprotein lipase (LPL). Seven residues, located on one face of a model alpha-helix spanning residues 59-75, are fully conserved in apoCII from ten different animal species. We have mutated these residues one by one. Substitution of Ala(59) by glycine, or Thr(62) and Gly(65) by alanine did not change the activation, indicating that these residues are outside the LPL-binding site. Replacement of Tyr(63), Ile(66), Asp(69), or Gln(70) by alanine lowered the affinity for LPL and the catalytic activity of the LPL-apoCII complex. For each residue several additional replacements were made. Most mutants retained some activating ability, but replacement of Tyr(63) by phenylalanine or tryptophan and Gln(70) by glutamate caused almost complete loss of activity. All mutants bound to liposomes with similar affinity as wild-type apoCII, and they also bound with similar affinity to LPL in the absence of hydrolyzable lipids. However, the inactive mutants did not compete with wild-type apoCII in the activation assay. Therefore, we conclude that the productive apoCII-LPL interaction may be dependent on substrate molecules. In summary, our data demonstrate that residues 63, 66, 69, and 70 are of special importance for the function of apoCII, but no single amino acid residue is absolutely crucial.


Subject(s)
Apolipoproteins C/physiology , Lipoprotein Lipase/metabolism , Apolipoprotein C-II , Apolipoproteins C/chemistry , Apolipoproteins C/genetics , Apolipoproteins C/isolation & purification , Base Sequence , Catalysis , Chromatography, High Pressure Liquid , Circular Dichroism , DNA Primers , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , Hydrolysis , Lipid Metabolism , Mutagenesis, Site-Directed , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
16.
Diabetes ; 50(12): 2779-85, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11723061

ABSTRACT

Apolipoprotein (APO) C1 is a 6.6-kDa protein present in plasma and associated with lipoproteins. Using hyperinsulinemic-euglycemic clamp tests, we previously found that in APOC1 transgenic mice, the whole-body insulin-mediated glucose uptake is increased concomitant with a decreased fatty acid uptake. These latter results are confirmed in the present study, showing that APOC1 transgenic mice exhibit a 50% reduction in the uptake of the fatty acid analog 15-(p-iodophenyl)-3-(R,S)-methyl pentadecanoic acid in white adipose tissue stores. We next investigated whether APOC1 overexpression can modulate the initiation and/or development of obesity and insulin resistance. When crossbred on the genetically obese ob/ob background, APOC1 transgenic mice were fully protected from the development of obesity compared with ob/ob only mice, as reflected by a strong reduction in body weight (21 +/- 4 vs. 44 +/- 7 g), total adipose tissue stores (15 +/- 3 vs. 25 +/- 3% body wt), and average adipocyte size (7,689 +/- 624 vs. 15,295 +/- 1,289 microm(2)). Although less pronounced, APOC1 overexpression also reduced body weight on a wild-type background, solely due to a reduction in adipose tissue. Furthermore, despite elevated plasma free fatty acid and triglyceride levels, APOC1 overexpression significantly improved insulin sensitivity in ob/ob mice, as demonstrated by a strong reduction in plasma glucose and insulin levels, as well as a better performance in the glucose tolerance test. In conclusion, a marked reduction in the uptake of fatty acids into adipocytes may underlie the protection from obesity and insulin resistance in transgenic mice overexpressing human APOC1.


Subject(s)
Apolipoproteins C/genetics , Gene Expression , Insulin Resistance/genetics , Obesity/genetics , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Apolipoprotein C-I , Apolipoproteins C/physiology , Blood Glucose/analysis , Cell Size , Cholesterol/blood , Crosses, Genetic , Fatty Acids/metabolism , Fatty Acids, Nonesterified/blood , Glucose Tolerance Test , Humans , Insulin/blood , Iodine Radioisotopes , Iodobenzenes/metabolism , Male , Mice , Mice, Obese , Mice, Transgenic , Obesity/pathology , Organ Size , Triglycerides/blood , Weight Loss/genetics
17.
Curr Opin Lipidol ; 12(3): 297-304, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11353333

ABSTRACT

Apolipoprotein (apo)C-I and apoC-III are constituents of HDL and of triglyceride-rich lipoproteins that slow the clearance of triglyceride-rich lipoproteins by a variety of mechanisms. ApoC-I is an inhibitor of lipoprotein binding to the LDL receptor, LDL receptor-related protein, and VLDL receptor. It also is the major plasma inhibitor of cholesteryl ester transfer protein, and appears to interfere directly with fatty acid uptake. ApoC-III also interferes with lipoprotein particle clearance, but its principal role is as an inhibitor of lipolysis, both through the biochemical inhibition of lipoprotein lipase and by interfering with lipoprotein binding to the cell-surface glycosaminoglycan matrix where lipolytic enzymes and lipoprotein receptors reside. Variation in the expression of apoC-III has been credibly documented to have an important role in hypertriglyceridemia. Variation in the expression of apoC-I may also be important for hypertriglyceridemia under certain circumstances.


Subject(s)
Apolipoproteins C/physiology , Lipoproteins/metabolism , Receptors, Immunologic/metabolism , Alleles , Animals , Apolipoprotein C-I , Apolipoprotein C-III , Apolipoproteins C/genetics , Gene Expression Regulation , Genetic Variation , Humans , Hypertriglyceridemia/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Receptors, LDL/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...