Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cancer Immunol Immunother ; 72(12): 4123-4144, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37853273

ABSTRACT

Increased prevalence of cancer in obese individuals is involved with dyslipidemia- induced chronic inflammation and immune suppression. Although apolipoprotein C-III (ApoC3)-transgenic mice (ApoC3TG mice) or poloxamer 407 (P407)-treated mice had hyperlipidemia, CD8+ T cells with upregulated antitumor activities were observed in ApoC3TG mice, and decreased CD8+ T cell activities were observed in P407-treated mice. Increased ApoC3 expression in hepatocellular carcinoma was associated with increased infiltration of CD8+ T cells and predicted survival. Recombinant ApoC3 had no direct effects on CD8+ T cells. The upregulation of CD8+ T cells in ApoC3TG mice was due to cross-talk with context cells, as indicated by metabolic changes and RNA sequencing results. In contrast to dendritic cells, the macrophages of ApoC3TG mice (macrophagesTG) displayed an activated phenotype and increased IL-1ß, TNF-α, and IL-6 production. Coculture with macrophagesTG increased CD8+ T cell function, and the adoptive transfer of macrophagesTG suppressed tumor progression in vivo. Furthermore, spleen tyrosine kinase (Syk) activation induced by TLR2/TLR4 cross-linking after ApoC3 ligation promoted cellular phospholipase A2 (cPLA2) activation, which in turn activated NADPH oxidase 2 (NOX2) to promote an alternative mode of inflammasome activation. Meanwhile, mitochondrial ROS produced by increased oxidative phosphorylation of free fatty acids facilitated the classical inflammasome activation, which exerted an auxiliary effect on inflammasome activation of macrophagesTG. Collectively, the increased antitumor activity of CD8+ T cells was mediated by the ApoC3-stimulated inflammasome activation of macrophages, and the mimetic ApoC3 peptides that can bind TLR2/4 could be a future strategy to target liver cancer.


Subject(s)
Inflammasomes , Neoplasms , Mice , Animals , Inflammasomes/metabolism , Apolipoprotein C-III/metabolism , Apolipoprotein C-III/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Toll-Like Receptor 2/metabolism , Macrophages/metabolism , Neoplasms/metabolism , Phospholipases A2, Cytosolic/metabolism , Phospholipases A2, Cytosolic/pharmacology , Mice, Inbred C57BL
2.
Biofactors ; 49(1): 153-172, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36039858

ABSTRACT

Apolipoprotein CIII (apoCIII) is increased in obesity-induced insulin resistance and type-2 diabetes. Emerging evidences support the advantages of small interfering RNAs (siRNAs) to target disease-causing genes. The aim of this study was to develop siRNAs for in vivo silencing of apoCIII and investigate if this results in metabolic improvements comparable to what we have seen using antisense oligonucelotides against apoCIII. Twenty-four siRNAs were synthesized and tested in a dual luciferase reporter assay. The eight best were selected, based on knockdown at 20 nM, and of these, two were selected based on IC50 values. In vivo experiments were performed in ob/ob mice, an obese animal model for diabetes. To determine the dose-dependency, efficacy, duration of effect and therapeutic dose we used a short protocol giving the apoCIII-siRNA mix for three days. To evaluate long-term metabolic effects mice were treated for three days, every second week for eight weeks. The siRNA mix effectively and selectively reduced expression of apoCIII in liver in vivo. Treatment had to be repeated every two weeks to maintain a suppression of apoCIII. The reduction of apoCIII resulted in increased LPL activity, lower triglycerides, reduced liver fat, ceased weight gain, enhanced insulin sensitivity, and improved glucose homeostasis. No off-target or side effects were observed during the eight-week treatment period. These results suggest that in vivo silencing of apoCIII with siRNA, is a promising approach with the potential to be used in the battle against obesity-induced metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Syndrome , Mice , Animals , Apolipoprotein C-III/genetics , Apolipoprotein C-III/metabolism , Apolipoprotein C-III/pharmacology , RNA, Small Interfering , Obesity
3.
Cardiovasc Pathol ; 41: 11-17, 2019.
Article in English | MEDLINE | ID: mdl-31004933

ABSTRACT

Atherosclerosis is a chronic inflammatory disease with lipid accumulation. Apolipoprotein C3 (APOC3), which is an important regulator of human lipid metabolism, is associated with multiple vascular mechanisms in atherosclerosis and proinflammatory responses. We have previously reported that the expression of inflammatory cytokine TNF-α is elevated in human endothelial cells (HUVECs) after APOC3 treatment. This study investigates the APOC3 signaling pathway involved in TNF-α-mediated expression of JAM-1 in HUVECs. Cultured HUVECs were exposed to APOC3 (50 µg/ml) for 16 h. Mechanistic studies were carried out by silencing TNF-α gene with lentiviral TNF-α-shRNA. Our study was based on the eight signaling pathway inhibitors to block the effect of APOC3 in HUVECs. The expression of JAM-1 was determined by qRT-PCR, Western blotting, and flow cytometry. IKK2 degradation and NF-κB p65 phosphorylation were determined by Western blotting. Our results showed that APOC3 significantly promoted the TNF-α-induced expression of JAM-1 in HUVECs. Inhibiting APOC3 reversed the TNF-α-induced overexpression of JAM-1. Moreover, APOC3 induced the expression of NF-κB p65 and degraded IκB. In conclusion, APOC3 promoted the expression of JAM-1 via the NF-κB, IKK2, and PI3K signaling pathway.


Subject(s)
Apolipoprotein C-III/pharmacology , Cell Adhesion Molecules/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , I-kappa B Kinase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Receptors, Cell Surface/metabolism , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cell Adhesion Molecules/genetics , Cells, Cultured , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Phosphorylation , Proteolysis , RNA Interference , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/enzymology , Tumor Necrosis Factor-alpha/genetics
4.
Cell Cycle ; 16(22): 2230-2238, 2017.
Article in English | MEDLINE | ID: mdl-28933987

ABSTRACT

Apolipoprotein CIII (ApoCIII) has been shown to be associated with the inflammatory response, but the mechanism of its inflammatory effects remains unclear. Because vascular endothelial cells (VECs) play a key role in the development of inflammation, the present study was performed to investigate inflammatory mechanisms induced by ApoCIII in VECs. In this study, we screened differentially expressed genes (DEGs) using RNA-sequencing. The results identified 390 up-regulated genes and 257 down-regulated genes. We performed GO functional classification and KEGG pathway analysis for DEGs. Analysis of sequencing data showed that 21 genes were related to the MAPK pathway. Finally, we investigated whether ApoCIII regulates the expression of pro-inflammatory cytokines via MAPK signaling pathway. The results showed that ApoCIII increased the expression levels of IL-6, TNF-α, VCAM-1 and ICAM-1 in VECs. ApoCIII activated the phosphorylation of ERK1/2 and p38 MAPK. An inhibitor of ERK1/2 and p38 MAPK decreased the protein levels of IL-6 and TNF-α. Our findings demonstrate that ApoCIII induces pro-inflammatory cytokine production in VECs via activation of ERK1/2 and p38 MAPK phosphorylation.


Subject(s)
Apolipoprotein C-III/pharmacology , Endothelial Cells/metabolism , Animals , Inflammation , Interleukin-6/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Signal Transduction/physiology , Swine , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
5.
Diabetologia ; 60(11): 2262-2273, 2017 11.
Article in English | MEDLINE | ID: mdl-28835988

ABSTRACT

AIM/HYPOTHESIS: Here, our aim was to examine whether VLDL and apolipoprotein (apo) CIII induce endoplasmic reticulum (ER) stress, inflammation and insulin resistance in skeletal muscle. METHODS: Studies were conducted in mouse C2C12 myotubes, isolated skeletal muscle and skeletal muscle from transgenic mice overexpressing apoCIII. RESULTS: C2C12 myotubes exposed to VLDL showed increased levels of ER stress and inflammatory markers whereas peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) and AMP-activated protein kinase (AMPK) levels were reduced and the insulin signalling pathway was attenuated. The effects of VLDL were also observed in isolated skeletal muscle incubated with VLDL. The changes caused by VLDL were dependent on extracellular signal-regulated kinase (ERK) 1/2 since they were prevented by the ERK1/2 inhibitor U0126 or by knockdown of this kinase by siRNA transfection. ApoCIII mimicked the effects of VLDL and its effects were also blocked by ERK1/2 inhibition, suggesting that this apolipoprotein was responsible for the effects of VLDL. Skeletal muscle from transgenic mice overexpressing apoCIII showed increased levels of some ER stress and inflammatory markers and increased phosphorylated ERK1/2 levels, whereas PGC-1α levels were reduced, confirming apoCIII effects in vivo. Finally, incubation of myotubes with a neutralising antibody against Toll-like receptor 2 abolished the effects of apoCIII on ER stress, inflammation and insulin resistance, indicating that the effects of apoCIII were mediated by this receptor. CONCLUSIONS/INTERPRETATION: These results imply that elevated VLDL in diabetic states can contribute to the exacerbation of insulin resistance by activating ERK1/2 through Toll-like receptor 2.


Subject(s)
Apolipoprotein C-III/pharmacology , Cholesterol, VLDL/pharmacology , Insulin/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Toll-Like Receptor 2/metabolism , Animals , Cell Line , Inflammation/drug therapy , Mice , Signal Transduction/drug effects
6.
Cell Mol Life Sci ; 71(7): 1289-303, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23949443

ABSTRACT

Apolipoprotein CIII (ApoCIII) not only serves as an inhibitor of triglyceride hydrolysis but also participates in diabetes-related pathological events such as hyperactivation of voltage-gated Ca(2+) (CaV) channels in the pancreatic ß cell. However, nothing is known about the molecular mechanisms whereby ApoCIII hyperactivates ß cell CaV channels. We now demonstrate that ApoCIII increased CaV1 channel open probability and density. ApoCIII enhanced whole-cell Ca(2+) currents and the CaV1 channel blocker nimodipine completely abrogated this enhancement. The effect of ApoCIII was not influenced by individual inhibition of PKA, PKC, or Src. However, combined inhibition of PKA, PKC, and Src counteracted the effect of ApoCIII, similar results obtained by coinhibition of PKA and Src. Moreover, knockdown of ß1 integrin or scavenger receptor class B type I (SR-BI) prevented ApoCIII from hyperactivating ß cell CaV channels. These data reveal that ApoCIII hyperactivates ß cell CaV1 channels through SR-BI/ß1 integrin-dependent coactivation of PKA and Src.


Subject(s)
Apolipoprotein C-III/pharmacology , CD36 Antigens/metabolism , Calcium Channels/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Insulin-Secreting Cells/metabolism , Integrin beta1/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Animals , Apolipoprotein C-III/metabolism , Apolipoprotein C-III/physiology , CD36 Antigens/genetics , Calcium/metabolism , Cells, Cultured , Electrophysiology , Female , Gene Knockdown Techniques , Integrin beta1/genetics , Integrin beta1/physiology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , RNA Interference , Up-Regulation
7.
Endocrinology ; 152(8): 3040-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21693679

ABSTRACT

Apolipoprotein CIII (ApoCIII) is mainly synthesized in the liver and is important for triglyceride metabolism. The plasma concentration of ApoCIII is elevated in patients with type 1 diabetes (T1D), and in vitro ApoCIII causes apoptosis in pancreatic ß-cells in the absence of inflammatory stress. Here, we investigated the effects of ApoCIII on function, signaling, and viability in intact rat pancreatic islets exposed to proinflammatory cytokines to model the intraislet inflammatory milieu in T1D. In contrast to earlier observations in mouse ß-cells, exposure of rat islets to ApoCIII alone (50 µg/ml) did not cause apoptosis. In the presence of the islet-cytotoxic cytokines IL-1ß + interferon-γ, ApoCIII reduced cytokine-mediated islet cell death and impairment of ß-cell function. ApoCIII had no effects on mitogen-activated protein kinases (c-Jun N-terminal kinase, p38, and ERK) and had no impact on IL-1ß-induced c-Jun N-terminal kinase activation. However, ApoCIII augmented cytokine-mediated nitric oxide (NO) production and inducible NO synthase expression. Further, ApoCIII caused degradation of the nuclear factor κB-inhibitor inhibitor of κB and stimulated Ser473-phosphorylation of the survival serine-threonine kinase Akt. Inhibition of the Akt signaling pathway by the phosphatidylinositol 3 kinase inhibitor LY294002 counteracted the antiapoptotic effect of ApoCIII on cytokine-induced apoptosis. We conclude that ApoCIII in the presence of T1D-relevant proinflammatory cytokines reduces rat pancreatic islet cell apoptosis via Akt.


Subject(s)
Apolipoprotein C-III/pharmacology , Apoptosis/drug effects , Cytokines/pharmacology , Insulin-Secreting Cells/drug effects , Proto-Oncogene Proteins c-akt/physiology , Animals , Calcium/metabolism , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Phosphorylation , Rats , Rats, Wistar , Signal Transduction
8.
Arterioscler Thromb Vasc Biol ; 30(11): 2242-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20829510

ABSTRACT

OBJECTIVE: To examine the direct effect of apolipoprotein CIII (apoCIII) on adipokine expressions that are involved in obesity, insulin resistance, or metabolic syndrome. METHODS AND RESULTS: ApoCIII in triglyceride-rich lipoproteins is elevated in patients with obesity, insulin resistance, or metabolic syndrome. Its level is also associated with proinflammatory adipokines. Fully differentiated mouse 3T3L1 adipocytes were incubated with apoCIII. ApoCIII activated nuclear factor κB of 3T3L1 adipocytes and induced the expression of monocyte chemoattractant protein (MCP) 1 and interleukin (IL) 6. ApoCIII also activated extracellular signal-regulated kinase and p38. Mitogen-activated protein kinase kinase (MEK)-1 inhibitor PD98059, but not p38 inhibitor SB203580, inhibited apoCIII-induced upregulation of MCP-1 and IL-6. Previously, it was shown that apoCIII activates proinflammatory signals through toll-like receptor (TLR) 2. TLR2-blocking antibody abolished activation of nuclear factor κB and extracellular signal-regulated kinase induced by apoCIII and inhibited apoCIII-induced upregulation of MCP-1 and IL-6. ApoCIII also reduced adiponectin expression of 3T3L1 adipocytes, which was recovered by TLR2-blocking antibody. ApoCIII induced the expression of MCP-1 and IL-6 in TLR2-overexpressed human embryonic kidney 293 cells but not wild-type human embryonic kidney 293 cells without TLR2. ApoCIII induced the expression of MCP-1 and IL-6 and decreased adiponectin expression in white adipose tissue of wild-type mice but not of TLR2-deficient mice in vivo. CONCLUSIONS: ApoCIII may activate extracellular signal-regulated kinase and nuclear factor kB through TLR2 and induce proinflammatory adipokine expression in vitro and in vivo. Thus, apoCIII links dyslipidemia to inflammation in adipocytes, which, in turn, may contribute to atherosclerosis.


Subject(s)
Adipocytes/metabolism , Apolipoprotein C-III/pharmacology , Chemokine CCL2/metabolism , Interleukin-6/metabolism , Lipoproteins, VLDL/pharmacology , Toll-Like Receptor 2/metabolism , Adipocytes/drug effects , Animals , Cells, Cultured , Humans , Male , Mice , NF-kappa B/metabolism
9.
Lipids Health Dis ; 8: 50, 2009 Nov 23.
Article in English | MEDLINE | ID: mdl-19930639

ABSTRACT

BACKGROUND: CETP is a plasma protein that modulates atherosclerosis risk through its HDL-cholesterol reducing action. The aim of this work was to examine the effect of the PPARalpha agonist, ciprofibrate, on the CETP gene expression, in the presence and absence of apolipoprotein (apo) CIII induced hypertriglyceridemia, and its impact on the HDL metabolism. RESULTS: Mice expressing apo CIII and/or CETP and non-transgenic littermates (CIII, CIII/CETP, CETP, non-Tg) were treated with ciprofibrate during 3 weeks. Drug treatment reduced plasma triglycerides (30-43%) and non-esterified fatty acids (19-47%) levels. Cholesterol (chol) distribution in plasma lipoprotein responses to ciprofibrate treatment was dependent on the genotypes. Treated CIII expressing mice presented elevation in VLDL-chol and reduction in HDL-chol. Treated CETP expressing mice responded with reduction in LDL-chol whereas in non-Tg mice the LDL-chol increased. In addition, ciprofibrate increased plasma post heparin lipoprotein lipase activity (1.3-2.1 fold) in all groups but hepatic lipase activity decreased in treated CETP and non-Tg mice. Plasma CETP activity and liver CETP mRNA levels were significantly increased in treated CIII/CETP and CETP mice (30-100%). Kinetic studies with 3H-cholesteryl ether (CEt) labelled HDL showed a 50% reduction in the 3H-CEt found in the LDL fraction in ciprofibrate treated compared to non-treated CETP mice. This means that 3H-CEt transferred from HDL to LDL was more efficiently removed from the plasma in the fibrate treated mice. Accordingly, the amount of 3H-CEt recovered in the liver 6 hours after HDL injection was increased by 35%. CONCLUSION: Together these data showed that the PPARalpha agonist ciprofibrate stimulates CETP gene expression and changes the cholesterol flow through the reverse cholesterol transport, increasing plasma cholesterol removal through LDL.


Subject(s)
Cholesterol Ester Transfer Proteins/genetics , Cholesterol/metabolism , Clofibric Acid/analogs & derivatives , Liver/metabolism , Animals , Apolipoprotein C-III/pharmacology , Biological Transport , Clofibric Acid/pharmacology , Fibric Acids , Gene Expression/drug effects , Hypertriglyceridemia/chemically induced , Mice , PPAR alpha/agonists
10.
Lipids Health Dis ; 8: 3, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19196457

ABSTRACT

BACKGROUND: Individuals with type 2 diabetes mellitus (T2DM) have elevated levels of circulating apolipoprotein CIII (apoCIII). ApoCIII plays an important role for plasma triglyceride levels and elevated levels of the apolipoprotein have been connected with dyslipidemia in T2DM subjects. In addition, apoCIII has been linked to enhanced beta-cell apoptosis. The present study was undertaken to investigate apoptotic mechanisms induced by the apolipoprotein. RESULTS: ApoCIII (10 microg/ml) enhanced apoptosis 2-fold in insulin-producing INS-1E cells after 24 hours exposure to the apolipoprotein. At this time point phosphorylation of mitogen activated protein kinase (MAPK) p38 had doubled but ERK1/2 and JNK were not activated. Instead, ERK1/2 showed rapid and transient phosphorylation (2-fold after 0.5 hour). No JNK phosphorylation was observed. In support of a role of activation of not only p38 but also ERK1/2 in apoCIII-induced apoptosis, inclusion of p38 inhibitor SB203580 (10 microM) or ERK1/2 inhibitor PD98059 (100 microM) normalized apoptosis. Whereas influx of Ca2+ was linked to apoCIII-induced ERK1/2 activation, pro-apoptotic protein CHOP/GADD of the unfolded protein response (UPR) was not affected by apoCIII. CONCLUSION: It is suggested that elevated circulating apoCIII levels may contribute to beta-cell apoptosis via activation of p38 and ERK1/2 in individuals with T2DM. Therapies aiming at normalizing levels of apoCIII could be beneficial not only for the function of the beta-cell but also for cardiovascular protection.


Subject(s)
Apolipoprotein C-III/pharmacology , Apoptosis/drug effects , Mitogen-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Rats , Transcription Factor CHOP/metabolism
11.
Biochemistry ; 47(31): 8186-94, 2008 Aug 05.
Article in English | MEDLINE | ID: mdl-18605697

ABSTRACT

Electronegative low-density lipoprotein (LDL(-)) is a minor LDL subfraction present in plasma with increased platelet-activating factor acetylhydrolase (PAF-AH) activity. This activity could be involved in the proinflammatory effects of LDL(-). Our aim was to study the presence of additional phospholipolytic activities in LDL(-). Total LDL was fractionated into electropositive (LDL(+)) and LDL(-) by anion-exchange chromatography, and phospholipolytic activities were measured by fluorometric methods. Phospholipolytic activity was absent in LDL(+) whereas LDL(-) presented activity against lysophosphatidylcholine (LPC, 82.4 +/- 34.9 milliunits/mg of apoB), sphingomyelin (SM, 53.3 +/- 22.5 milliunits/mg of apoB), and phosphatidylcholine (PC, 25.7 +/- 4.3 milliunits/mg of apoB). LDL(-), but not LDL(+), presented spontaneous self-aggregation at 37 degrees C in parallel to phospholipid degradation. This was observed in the absence of lipid peroxidation and suggests the involvement of phospholipolytic activity in self-aggregation of LDL(-). Phospholipolytic activity was not due to PAF-AH, apoE, or apoC-III and was not increased in LDL(+) modified by Cu (2+) oxidation, acetylation, or secretory phospholipase A 2 (PLA 2). However, LDL(-) efficiently degraded phospholipids of lipoproteins enriched in LPC, such as oxidized LDL or PLA 2-LDL, but not native or acetylated LDL. This finding supports that LPC is the best substrate for LDL(-)-associated phospholipolytic activity. These results reveal novel properties of LDL(-) that could play a significant role in its atherogenic properties.


Subject(s)
Lipolysis/physiology , Lipoproteins, LDL/metabolism , Phospholipids/metabolism , Anion Exchange Resins/chemistry , Apolipoprotein C-III/pharmacology , Apolipoproteins E/pharmacology , Bridged-Ring Compounds/pharmacology , Chromatography, Ion Exchange , Enzyme Activation/drug effects , Hydrogen-Ion Concentration , Lipolysis/drug effects , Lipoproteins, LDL/chemistry , Lysophosphatidylcholines/chemistry , Lysophosphatidylcholines/metabolism , Magnesium/metabolism , Norbornanes , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phospholipids/chemistry , Sphingomyelins/chemistry , Sphingomyelins/metabolism , Sulfones/pharmacology , Thiocarbamates , Thiones/pharmacology , Time Factors
12.
Circulation ; 118(7): 731-42, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18663085

ABSTRACT

BACKGROUND: Apolipoprotein CIII (apoCIII) is a component of some triglyceride-rich very-low-density and low-density lipoprotein and is elevated in dyslipidemia with insulin resistance and the metabolic syndrome. We previously reported that apoCIII directly activates proinflammatory and atherogenic signaling in vascular endothelial cells through protein kinase C-beta (PKCbeta). Because PKCbeta impairs the response of vascular endothelial cells to insulin, we tested the hypothesis that apoCIII affects insulin signaling in vascular endothelial cells and its function in vitro and in vivo. METHODS AND RESULTS: ApoCIII inhibited insulin-induced tyrosine phosphorylation of insulin receptor substrate 1 (IRS-1), decreasing phosphatidylinositol 3-kinase (PI3K)/Akt activation in human umbilical vein endothelial cells. These effects of apoCIII led to reduced endothelial nitric oxide synthase (eNOS) activation and NO release into the media. ApoCIII activated PKCbeta in human umbilical vein endothelial cells, resulting in IRS-1 dysfunction via serine phosphorylation. ApoCIII also activated mitogen-activated protein kinase through PKCbeta. The impaired insulin signaling was restored by PKCbeta inhibitor or MEK1 inhibitor. ApoCIII-rich very-low-density lipoprotein and apoCIII impaired insulin signaling in the aorta of C57BL/6J mice and in human umbilical vein endothelial cells, which was recovered by PKCbeta inhibitor. They also inhibited endothelium-dependent relaxation of the aortas of C57BL/6J mice. In summary, apoCIII in very-low-density lipoprotein impaired insulin stimulation of NO production by vascular endothelium and induced endothelial dysfunction in vivo. This adverse effect of apoCIII was mediated by its activation of PKCbeta, which inhibits the IRS-1/PI3K/Akt/eNOS pathway. CONCLUSIONS: Our results suggest that apoCIII is a crucial link between dyslipidemia and insulin resistance in vascular endothelial cells with consequential deleterious effects on their atheroprotective functions.


Subject(s)
Apolipoprotein C-III/metabolism , Endothelial Cells/metabolism , Hyperlipidemias/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/physiopathology , Apolipoprotein C-III/pharmacology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Humans , Hyperlipidemias/physiopathology , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Insulin Resistance/physiology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C/metabolism , Protein Kinase C beta , Proto-Oncogene Proteins c-akt/metabolism , Vasodilation/drug effects , Vasodilation/physiology
13.
J Lipid Res ; 49(5): 1048-55, 2008 May.
Article in English | MEDLINE | ID: mdl-18263930

ABSTRACT

Apolipoprotein E2 (apoE2)-associated hyperlipidemia is characterized by a disturbed clearance of apoE2-enriched VLDL remnants. Because excess apoE2 inhibits LPL-mediated triglyceride (TG) hydrolysis in vitro, we investigated whether direct or indirect stimulation of LPL activity in vivo reduces the apoE2-associated hypertriglyceridemia. Here, we studied the role of LPL and two potent modifiers, the LPL inhibitor apoC-III and the LPL activator apoA-V, in APOE2-knockin (APOE2) mice. Injection of heparin in APOE2 mice reduced plasma TG by 53% and plasma total cholesterol (TC) by 18%. Adenovirus-mediated overexpression of LPL reduced plasma TG by 85% and TC by 40%. Both experiments indicate that the TG in apoE2-enriched particles is a suitable substrate for LPL. Indirect activation of LPL activity via deletion of Apoc3 in APOE2 mice did not affect plasma TG levels, whereas overexpression of Apoa5 in APOE2 mice did reduce plasma TG by 81% and plasma TC by 41%. In conclusion, the hypertriglyceridemia in APOE2 mice can be ameliorated by the direct activation of LPL activity. Indirect activation of LPL via overexpression of apoA-V does, whereas deletion of apoC-III does not, affect the plasma TGs in APOE2 mice. These data indicate that changes in apoA-V levels have a dominant effect over changes in apoC-III levels in the improvement of APOE2-associated hypertriglyceridemia.


Subject(s)
Apolipoprotein C-III/pharmacology , Apolipoprotein E2/physiology , Apolipoproteins/pharmacology , Hypertriglyceridemia/blood , Adenoviridae , Animals , Apolipoprotein A-V , Apolipoprotein E2/deficiency , Apolipoprotein E2/genetics , Gene Transfer Techniques , Lipids/blood , Lipoprotein Lipase/blood , Male , Mice , Mice, Knockout
15.
Circ Res ; 100(3): 381-90, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17234968

ABSTRACT

High levels of triglyceride-rich lipoproteins (TGRLs) in blood are linked to development of atherosclerosis, yet the mechanisms by which these particles initiate inflammation of endothelium are unknown. TGRL isolated from human plasma during the postprandial state was examined for its capacity to bind to cultured human aortic endothelial cells (HAECs) and alter the acute inflammatory response to tumor necrosis factor-alpha. HAECs were repetitively incubated with dietary levels of freshly isolated TGRL for 2 hours per day for 1 to 3 days to mimic postprandial lipidemia. TGRL induced membrane upregulation of the low-density lipoprotein family receptors LRP and LR11, which was inhibited by the low-density lipoprotein receptor-associated protein-1. TGRLs alone did not elicit inflammation in HAECs but enhanced the inflammatory response via a 10-fold increase in sensitivity to cytokine stimulation. This was reflected by increased mitogen-activated protein kinase activation, nuclear translocation of NF-kappaB, amplified expression of endothelial selectin and VCAM-1, and a subsequent increase in monocyte-specific recruitment under shear flow as quantified in a microfabricated vascular mimetic device.


Subject(s)
Aortic Diseases/etiology , Arteriosclerosis/etiology , Arteritis/etiology , Dietary Fats/adverse effects , Endothelial Cells/drug effects , Hypertriglyceridemia/complications , LDL-Receptor Related Proteins/metabolism , Lipoproteins, HDL/toxicity , Lipoproteins, LDL/toxicity , Lipoproteins, VLDL/toxicity , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Membrane Transport Proteins/metabolism , Receptors, LDL/metabolism , Triglycerides/toxicity , Tumor Necrosis Factor-alpha/pharmacology , Aorta , Apolipoprotein C-III/metabolism , Apolipoprotein C-III/pharmacology , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chylomicrons/blood , Dietary Fats/administration & dosage , E-Selectin/biosynthesis , E-Selectin/genetics , Endocytosis , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Fat Emulsions, Intravenous/pharmacology , Gene Expression Regulation/drug effects , Humans , Hypertriglyceridemia/blood , Hypoglycemia , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , LDL-Receptor Related Protein-Associated Protein/pharmacology , LDL-Receptor Related Proteins/drug effects , Leukocytes/cytology , Leukocytes/drug effects , Lipopolysaccharides/pharmacology , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Low Density Lipoprotein Receptor-Related Protein-1/drug effects , Membrane Transport Proteins/drug effects , Models, Cardiovascular , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/metabolism , Oxidative Stress , Receptors, LDL/drug effects , Rheology , Signal Transduction/drug effects , Triglycerides/blood , Tumor Necrosis Factor-alpha/physiology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...