Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 170
Filter
1.
Article in English | MEDLINE | ID: mdl-34610468

ABSTRACT

Hydrolysis of VLDL triacylglycerol (TG) by lipoprotein lipase (LpL) is a major step in energy metabolism and VLDL-to-LDL maturation. Most functional LpL is anchored to the vascular endothelium, yet a small amount circulates on TG-rich lipoproteins. As circulating LpL has low catalytic activity, its role in VLDL remodeling is unclear. We use pre-heparin plasma and heparin-sepharose affinity chromatography to isolate VLDL fractions from normolipidemic, hypertriglyceridemic, or type-2 diabetic subjects. LpL is detected only in the heparin-bound fraction. Transient binding to heparin activates this VLDL-associated LpL, which hydrolyses TG, leading to gradual VLDL remodeling into IDL/LDL and HDL-size particles. The products and the timeframe of this remodeling closely resemble VLDL-to-LDL maturation in vivo. Importantly, the VLDL fraction that does not bind heparin is not remodeled. This relatively inert LpL-free VLDL is rich in TG and apoC-III, poor in apoE and apoC-II, shows impaired functionality as a substrate for the exogenous LpL or CETP, and likely has prolonged residence time in blood, which is expected to promote atherogenesis. This non-bound VLDL fraction increases in hypertriglyceridemia and in type-2 diabetes but decreases upon diabetes treatment that restores the glycemic control. In stark contrast, heparin binding by LDL increases in type-2 diabetes triggering pro-atherogenic LDL modifications. Therefore, the effects of heparin binding are associated negatively with atherogenesis for VLDL but positively for LDL. Collectively, the results reveal that binding to glycosaminoglycans initiates VLDL remodeling by circulating LpL, and suggest heparin binding as a marker of VLDL functionality and a readout for treatment of metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Hypertriglyceridemia/genetics , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/genetics , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/metabolism , Energy Metabolism/genetics , Heparin/genetics , Heparin/metabolism , Humans , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Lipoproteins, LDL/genetics , Triglycerides/genetics
2.
J Biol Chem ; 296: 100312, 2021.
Article in English | MEDLINE | ID: mdl-33482195

ABSTRACT

Elevated plasma triglycerides are a risk factor for coronary artery disease, which is the leading cause of death worldwide. Lipoprotein lipase (LPL) reduces triglycerides in the blood by hydrolyzing them from triglyceride-rich lipoproteins to release free fatty acids. LPL activity is regulated in a nutritionally responsive manner by macromolecular inhibitors including angiopoietin-like proteins 3 and 4 (ANGPTL3 and ANGPTL4). However, the mechanism by which ANGPTL3 inhibits LPL is unclear, in part due to challenges in obtaining pure protein for study. We used a new purification protocol for the N-terminal domain of ANGPTL3, removing a DNA contaminant, and found DNA-free ANGPTL3 showed enhanced inhibition of LPL. Structural analysis showed that ANGPTL3 formed elongated, flexible trimers and hexamers that did not interconvert. ANGPTL4 formed only elongated flexible trimers. We compared the inhibition of ANGPTL3 and ANGPTL4 using human very-low-density lipoproteins as a substrate and found both were noncompetitive inhibitors. The inhibition constants for the trimeric ANGPTL3 (7.5 ± 0.7 nM) and ANGPTL4 (3.6 ± 1.0 nM) were only 2-fold different. Heparin has previously been reported to interfere with ANGPTL3 binding to LPL, so we questioned if the negatively charged heparin was acting in a similar fashion to the DNA contaminant. We found that ANGPTL3 inhibition is abolished by binding to low-molecular-weight heparin, whereas ANGPTL4 inhibition is not. Our data show new similarities and differences in how ANGPTL3 and ANGPTL4 regulate LPL and opens new avenues of investigating the effect of heparin on LPL inhibition by ANGPTL3.


Subject(s)
Angiopoietin-Like Protein 4/chemistry , Angiopoietin-like Proteins/chemistry , Coronary Artery Disease/genetics , Lipoprotein Lipase/chemistry , Protein Conformation , Angiopoietin-Like Protein 3 , Angiopoietin-Like Protein 4/genetics , Angiopoietin-Like Protein 4/ultrastructure , Angiopoietin-like Proteins/genetics , Angiopoietin-like Proteins/ultrastructure , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Heparin/pharmacology , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/ultrastructure , Lipoproteins, VLDL/chemistry , Lipoproteins, VLDL/genetics , Protein Binding/drug effects , Substrate Specificity , Triglycerides/blood
3.
JCI Insight ; 5(24)2020 12 17.
Article in English | MEDLINE | ID: mdl-33170809

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic lipid accumulation. The transmembrane 6 superfamily member 2 (TM6SF2) E167K genetic variant associates with NAFLD and with reduced plasma triglyceride levels in humans. However, the molecular mechanisms underlying these associations remain unclear. We hypothesized that TM6SF2 E167K affects hepatic very low-density lipoprotein (VLDL) secretion and studied the kinetics of apolipoprotein B100 (apoB100) and triglyceride metabolism in VLDL in homozygous subjects. In 10 homozygote TM6SF2 E167K carriers and 10 matched controls, we employed stable-isotope tracer and compartmental modeling techniques to determine apoB100 and triglyceride kinetics in the 2 major VLDL subfractions: large triglyceride-rich VLDL1 and smaller, less triglyceride-rich VLDL2. VLDL1-apoB100 production was markedly reduced in homozygote TM6SF2 E167K carriers compared with controls. Likewise, VLDL1-triglyceride production was 35% lower in the TM6SF2 E167K carriers. In contrast, the direct production rates for VLDL2-apoB100 and triglyceride were not different between carriers and controls. In conclusion, the TM6SF2 E167K genetic variant was linked to a specific reduction in hepatic secretion of large triglyceride-rich VLDL1. The impaired secretion of VLDL1 explains the reduced plasma triglyceride concentration and provides a basis for understanding the lower risk of cardiovascular disease associated with the TM6SF2 E167K genetic variant.


Subject(s)
Lipoproteins, VLDL/metabolism , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Apolipoprotein B-100/metabolism , Female , Genetic Predisposition to Disease , Humans , Lipase/metabolism , Lipid Metabolism/genetics , Lipids/genetics , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/genetics , Liver/metabolism , Liver/pathology , Male , Membrane Proteins/metabolism , Middle Aged , Non-alcoholic Fatty Liver Disease/metabolism , Polymorphism, Single Nucleotide/genetics , Triglycerides/metabolism
4.
Circ Res ; 127(11): 1347-1361, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32912065

ABSTRACT

RATIONALE: Single-nucleotide polymorphisms near the ILRUN (inflammation and lipid regulator with ubiquitin-associated-like and NBR1 [next to BRCA1 gene 1 protein]-like domains) gene are genome-wide significantly associated with plasma lipid traits and coronary artery disease (CAD), but the biological basis of this association is unknown. OBJECTIVE: To investigate the role of ILRUN in plasma lipid and lipoprotein metabolism. METHODS AND RESULTS: ILRUN encodes a protein that contains a ubiquitin-associated-like domain, suggesting that it may interact with ubiquitinylated proteins. We generated mice globally deficient for Ilrun and found they had significantly lower plasma cholesterol levels resulting from reduced liver lipoprotein production. Liver transcriptome analysis uncovered altered transcription of genes downstream of lipid-related transcription factors, particularly PPARα (peroxisome proliferator-activated receptor alpha), and livers from Ilrun-deficient mice had increased PPARα protein. Human ILRUN was shown to bind to ubiquitinylated proteins including PPARα, and the ubiquitin-associated-like domain of ILRUN was found to be required for its interaction with PPARα. CONCLUSIONS: These findings establish ILRUN as a novel regulator of lipid metabolism that promotes hepatic lipoprotein production. Our results also provide functional evidence that ILRUN may be the casual gene underlying the observed genetic associations with plasma lipids at 6p21 in human.


Subject(s)
Hepatocytes/metabolism , Lipoproteins/blood , Liver/metabolism , Animals , Blood Glucose/metabolism , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Cholesterol, HDL/blood , Cholesterol, HDL/genetics , Gene Expression Regulation , Glucose Intolerance/blood , Glucose Intolerance/genetics , HEK293 Cells , Humans , Lipoproteins/genetics , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Protein Binding , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Transcriptome , Triglycerides/blood , Triglycerides/genetics , Ubiquitination
5.
Curr Opin Lipidol ; 31(3): 132-139, 2020 06.
Article in English | MEDLINE | ID: mdl-32332433

ABSTRACT

PURPOSE OF REVIEW: To critically appraise new insights into the biology of remnant lipoproteins and their putative role in the pathophysiology of atherosclerotic cardiovascular disease, and to compare the atherogenicity of remnant particles with that of low-density lipoproteins (LDL). RECENT FINDINGS: New in-vivo stable isotope tracer studies of the kinetics of apoB48 and apoB100-containing lipoproteins in postprandial conditions have revealed that apoB48-containing very low-density lipoproteins (VLDL) accumulated markedly in hypertriglyceridemic patients. These intestinally-derived particles were cleared slowly, and represented up to 25% of circulating VLDL; as part of the remnant particle population, they may increase cardiovascular risk. Importantly, the PCSK9 inhibitor, evolocumab, was shown to reduce remnant levels (-29%) during the postprandial period in diabetic patients on statin therapy - an effect which may be additive to that of LDL-cholesterol reduction in conferring cardiovascular benefit. In recent Mendelian randomization studies, the effect of lowering triglyceride-rich lipoproteins or LDL-cholesterol translated to similar clinical benefit per unit of apoB. Finally, in randomized trials involving statin-treated patients with atherosclerotic cardiovascular disease, remnant cholesterol levels were associated with coronary atheroma progression independently of LDL-cholesterol. SUMMARY: Overall, data from observational studies in large cohorts, Mendelian randomization studies, meta-regression analyses, and post-hoc analyses of randomized trials are consistent with the contention that remnants are highly atherogenic particles and contribute to the atherosclerotic burden in an equivalent manner to that of LDL.


Subject(s)
Apolipoprotein B-100/genetics , Atherosclerosis/genetics , Cardiovascular Diseases/genetics , Proprotein Convertase 9/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Humans , Isotope Labeling , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/metabolism , PCSK9 Inhibitors
6.
J Cell Physiol ; 235(9): 6246-6256, 2020 09.
Article in English | MEDLINE | ID: mdl-32012270

ABSTRACT

To evaluate the effects of fatty acids on endoplasmic reticulum (ER) stress, oxidative stress, and lipid damage. We treated BRL3A rat liver cells with, linoleic (LA), linolenic, oleic (OA), palmitic (PA), palmitoleic (POA), or stearic (SA) acid for 12 hr. The characteristics of cell lipid deposition, oxidative stress indexes, ER stress markers, nuclear factor κB p65 (NF-κB p65), lipid synthesis and transport regulators, and cholesterol metabolism regulators were analyzed. Endoplasmic chaperones like glucose-regulated protein 78, CCAAT-enhancer-binding protein, NF-κB p65, hydrogen peroxide, and malonaldehyde in PA- and SA-treated cells were significantly higher than in other treated cells. Deposition of fatty acids especially LA and POA were significantly increased than in other treated cells. De novo lipogenesis regulators sterol regulatory element-binding protein 1c, fatty acid synthase, and acetyl-coenzyme A carboxylase 1 (ACC1) expression were significantly increased in all fatty acid stimulation groups, and PA- and SA-treated cells showed lower p-ACC1 expression and higher scd1 expression than other fatty acid groups. Very low-density lipoprotein synthesis and apolipoprotein B100 expression in free fatty acids treated cells were significantly lower than control. PA, SA, OA, and POA had shown significantly increased cholesterol synthesis than other treated cells. PA and SA showed the lower synthesis of cytochrome P7A1 and total bile acids than other fatty acids treated cells. Excess of saturated fatty acids led to severe ER and oxidative stress. Excess unsaturated fatty acids led to increased lipid deposition in cultured hepatocytes. A balanced fatty acid intake is needed to maintain lipid homeostasis.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Fatty Acids/pharmacology , Fatty Liver/drug therapy , Lipogenesis/genetics , Acetyl-CoA Carboxylase/genetics , Animals , Cells, Cultured , Fatty Acid Synthases/genetics , Fatty Acids/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Linoleic Acid/pharmacology , Lipid Metabolism/drug effects , Lipids/biosynthesis , Lipids/genetics , Lipogenesis/drug effects , Lipoproteins, VLDL/genetics , Liver/drug effects , Liver/metabolism , Oleic Acid/pharmacology , Oxidative Stress/drug effects , Palmitic Acid/pharmacology , Rats , Stearic Acids/pharmacology , alpha-Linolenic Acid/pharmacology
7.
J Clin Lipidol ; 14(1): 77-87, 2020.
Article in English | MEDLINE | ID: mdl-31917184

ABSTRACT

BACKGROUND: Monoclonal antibodies to proprotein convertase subtilisin/kexin type 9 (PCSK9) significantly lower the levels of low-density lipoprotein and very-low-density lipoproteins (VLDL), but their effect on postprandial lipoprotein metabolism in dyslipidemic subjects is unclear. OBJECTIVE: This study aimed to investigate the effects of evolocumab on postprandial lipid responses, ectopic fat depots, whole-body cholesterol synthesis, hepatic lipogenesis, and fat oxidation in patients with type II diabetes. METHODS: The trial was a single-phase, nonrandomized study of 12-week treatment with evolocumab 140 mg subcutaneously every 2 weeks in 15 patients with type II diabetes on background statin therapy. Cardiometabolic responses to a high-fat mixed meal were assessed before and at the end of the intervention period. RESULTS: Evolocumab treatment reduced significantly postprandial rises in plasma total triglyceride (by 21%; P < .0001) and VLDL1 triglyceride (by 15%; P = .018), but the increase in chylomicron triglyceride after the meal was not significantly perturbed (P = .053). There were reduced postprandial responses in plasma total apolipoprotein C-III (by 14%; P < .0001) and apolipoprotein B-48 concentration (by 17%; P = .0046) and in "remnant-like particles" cholesterol (by 29%; P < .0001) on the PCSK9 inhibitor. Treatment reduced the steady-state (ie, fasting and postprandial) concentrations of VLDL2 cholesterol by 50% (P < .0001) and VLDL2 triglyceride by 29% (P < .0001), in addition to the 78% reduction of low-density lipoprotein cholesterol (P < .001). The changes in apolipoprotein C-III associated significantly with reduction in postprandial responses of remnant-like particles cholesterol and triglyceride-rich lipoprotein cholesterol. Evolocumab therapy did not influence liver fat accumulation, hepatic de novo lipogenesis, or fasting ß-hydroxybutyrate but did increase total body cholesterol synthesis (P < .01). CONCLUSION: Evolocumab treatment improved postprandial responses of triglyceride-rich lipoproteins and measures of cholesterol-enriched remnant particles in type II diabetic subjects. These results indicate that postprandial phenomena need to be taken into account in assessing the full range of actions of PCSK9 inhibitors in dyslipidemic individuals.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Lipoproteins, VLDL/genetics , Proprotein Convertase 9/blood , Adult , Cholesterol/blood , Chylomicrons/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Female , Humans , Lipoproteins/blood , Lipoproteins, VLDL/blood , Male , Middle Aged , PCSK9 Inhibitors , Postprandial Period , Proprotein Convertase 9/genetics , Triglycerides/blood
8.
Lipids Health Dis ; 18(1): 224, 2019 Dec 19.
Article in English | MEDLINE | ID: mdl-31856839

ABSTRACT

BACKGROUND: APOC3 is important in lipid transport and metabolism with limited studies reporting genetic sequence variations in specific ethnic groups. The present study aimed to analyze the full APOC3 sequence among Kuwaiti Arabs and test the association of selected variants with lipid levels and BMI. METHODS: Variants were identified by Sanger sequencing the entire APOC3 gene in 100 Kuwaiti Arabs. Variants and their genotypes were fully characterized and used to construct haplotype blocks. Four variants (rs5128, rs2854117, rs2070668, KUAPOC3N3 g.5196 A > G) were selected for testing association with serum lipid levels and BMI in a cohort (n = 733). RESULTS: APOC3 sequence (4.3 kb) of a Kuwaiti Arab was deposited in Genbank (accession number KJ437193). Forty-two variants including 3 novels were identified including an "A" insertion at genomic positions 116,700,599-116,700,600 (promoter region) and two substitutions in intron 1 at genomic positions 116,700,819 and 116,701,159. Only three variants, (rs5128, rs2854117, and rs2070668) were analyzed for association of which rs5128 showed a trend for association with increased BMI, TG and VLDL levels that was further investigated using multivariate analysis. A significant association of rs5128 with BMI (p <  0.05) was observed following a dominant genetic model with increased risk by an OR of 4.022 (CI: 1.13-14.30). CONCLUSION: The present study is the first to report sequence analysis of APOC3 in an Arab ethnic group. This study supports the inclusion of rs5128 as a marker for assessing genetic risk to dyslipidemia and obesity and the inclusion of the novel variant g.5196 A > G for population stratification of Arabs.


Subject(s)
Apolipoprotein C-III/genetics , Body Mass Index , Genetic Association Studies , Genetic Predisposition to Disease , Adolescent , Adult , Aged , Arabs/genetics , Female , Genotype , Humans , Lipid Metabolism/genetics , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , Triglycerides/blood , Triglycerides/genetics , Young Adult
9.
Nat Commun ; 10(1): 4788, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31636271

ABSTRACT

Genetic studies of metabolites have identified thousands of variants, many of which are associated with downstream metabolic and obesogenic disorders. However, these studies have relied on univariate analyses, reducing power and limiting context-specific understanding. Here we aim to provide an integrated perspective of the genetic basis of metabolites by leveraging the Finnish Metabolic Syndrome In Men (METSIM) cohort, a unique genetic resource which contains metabolic measurements, mostly lipids, across distinct time points as well as information on statin usage. We increase effective sample size by an average of two-fold by applying the Covariates for Multi-phenotype Studies (CMS) approach, identifying 588 significant SNP-metabolite associations, including 228 new associations. Our analysis pinpoints a small number of master metabolic regulator genes, balancing the relative proportion of dozens of metabolite levels. We further identify associations to changes in metabolic levels across time as well as genetic interactions with statin at both the master metabolic regulator and genome-wide level.


Subject(s)
Genetic Pleiotropy , Metabolic Syndrome/genetics , Metabolome/genetics , Aged , Amino Acids/genetics , Amino Acids/metabolism , Cohort Studies , Fatty Acids/genetics , Fatty Acids/metabolism , Gene Regulatory Networks , Genome-Wide Association Study , Humans , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Lipoproteins, IDL/genetics , Lipoproteins, IDL/metabolism , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/metabolism , Magnetic Resonance Spectroscopy , Male , Middle Aged , Polymorphism, Single Nucleotide
10.
J Clin Invest ; 129(11): 4885-4900, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31408437

ABSTRACT

Deciphering novel pathways regulating liver lipid content has profound implications for understanding the pathophysiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Recent evidence suggests that the nuclear envelope is a site of regulation of lipid metabolism but there is limited appreciation of the responsible mechanisms and molecular components within this organelle. We showed that conditional hepatocyte deletion of the inner nuclear membrane protein lamina-associated polypeptide 1 (LAP1) caused defective VLDL secretion and steatosis, including intranuclear lipid accumulation. LAP1 binds to and activates torsinA, an AAA+ ATPase that resides in the perinuclear space and continuous main ER. Deletion of torsinA from mouse hepatocytes caused even greater reductions in VLDL secretion and profound steatosis. Both of these mutant mouse lines developed hepatic steatosis and subsequent steatohepatitis on a regular chow diet in the absence of whole-body insulin resistance or obesity. Our results establish an essential role for the nuclear envelope-localized torsinA-LAP1 complex in hepatic VLDL secretion and suggest that the torsinA pathway participates in the pathophysiology of nonalcoholic fatty liver disease.


Subject(s)
Carrier Proteins/metabolism , Hepatocytes/metabolism , Lipoproteins, VLDL/metabolism , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Nuclear Envelope/metabolism , Animals , Carrier Proteins/genetics , Hepatocytes/pathology , Lipid Metabolism , Lipoproteins, VLDL/genetics , Membrane Proteins/genetics , Mice , Mice, Knockout , Molecular Chaperones/genetics , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Nuclear Envelope/genetics , Nuclear Envelope/pathology
11.
J Biol Chem ; 294(21): 8577-8591, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30971429

ABSTRACT

Enhancer of zeste homolog 2 (EZH2), an epigenetic regulator that plays a key role in cell differentiation and oncogenesis, was reported to promote adipogenic differentiation in vitro by catalyzing trimethylation of histone 3 lysine 27. However, inhibition of EZH2 induced lipid accumulation in certain cancer and hepatocyte cell lines. To address this discrepancy, we investigated the role of EZH2 in adipogenic differentiation and lipid metabolism using primary human and mouse preadipocytes and adipose-specific EZH2 knockout (KO) mice. We found that the EZH2-selective inhibitor GSK126 induced lipid accumulation in human adipocytes, without altering adipocyte differentiation marker gene expression. Moreover, adipocyte-specific EZH2 KO mice, generated by crossing EZH2 floxed mice with adiponectin-Cre mice, displayed significantly increased body weight, adipose tissue mass, and adipocyte cell size and reduced very low-density lipoprotein (VLDL) levels, as compared with littermate controls. These phenotypic alterations could not be explained by differences in feeding behavior, locomotor activity, metabolic energy expenditure, or adipose lipolysis. In addition, human adipocytes treated with either GSK126 or vehicle exhibited comparable rates of glucose-stimulated triglyceride accumulation and fatty acid uptake. Mechanistically, lipid accumulation induced by GSK126 in adipocytes was lipoprotein-dependent, and EZH2 inhibition or gene deletion promoted lipoprotein-dependent lipid uptake in vitro concomitant with up-regulated apolipoprotein E (ApoE) gene expression. Deletion of ApoE blocked the effects of GSK126 to promote lipoprotein-dependent lipid uptake in murine adipocytes. Collectively, these results indicate that EZH2 inhibition promotes lipoprotein-dependent lipid accumulation via inducing ApoE expression in adipocytes, suggesting a novel mechanism of lipid regulation by EZH2.


Subject(s)
Adipocytes/metabolism , Apolipoproteins E/metabolism , Cell Differentiation , Enhancer of Zeste Homolog 2 Protein/metabolism , Lipogenesis , Lipolysis , Adipocytes/cytology , Animals , Apolipoproteins E/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/metabolism , Mice , Up-Regulation
12.
Sci Rep ; 9(1): 5572, 2019 04 03.
Article in English | MEDLINE | ID: mdl-30944368

ABSTRACT

Lipoprotein lipase (LPL) is a rate-limiting enzyme for the hydrolysis of triglycerides (TG). Hundreds of genetic variants including single nucleotide polymorphisms have been identified across the 30Kb gene locus on chromosome 8q22. Several of these variants have been demonstrated to have genetic association with lipid level variation but many remain unresolved. Controversial reports on the genetic association of variants among different populations pose a challenge to which variants are informative. This study aimed to investigate "common" LPL variants (rs1121923, rs258, rs328, rs13702) and their possible role in plasma lipid level. Genotyping was performed using Realtime PCR. Based on the observed genotypes, the minor allele frequencies were A: 0.065 for rs1121923; C: 0.379 for rs258; G: 0.087 for rs328 and C: 0.337 for rs13702. Using linear regression, a lowering effect of rs1121923 (p = 0.024) on TG levels (-0.14 B coefficient: CI: -0.27--0.019) and rs258 (p = 0.013) on VLDL levels (B: -0.046; CI: -0.082--0.009) was observed indicating a "protective" role for the two variants. Moreover, the findings indicate the potential for including rs1121923 and rs258 in diagnostic panels for use as an estimator of "risk" scores for dyslipidemia.


Subject(s)
Lipoprotein Lipase/genetics , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/genetics , Polymorphism, Single Nucleotide/genetics , Triglycerides/blood , Triglycerides/genetics , Adolescent , Adult , Aged , Female , Gene Frequency/genetics , Genetic Association Studies/methods , Genotype , Humans , Male , Middle Aged , Young Adult
13.
FASEB J ; 33(7): 8666-8687, 2019 07.
Article in English | MEDLINE | ID: mdl-31002535

ABSTRACT

We recently described myonectin (also known as erythroferrone) as a novel skeletal muscle-derived myokine with metabolic functions. Here, we use a genetic mouse model to determine myonectin's requirement for metabolic homeostasis. Female myonectin-deficient mice had larger gonadal fat pads and developed mild insulin resistance when fed a high-fat diet (HFD) and had reduced food intake during refeeding after an unfed period but were otherwise indistinguishable from wild-type littermates. Male mice lacking myonectin, however, had reduced physical activity when fed ad libitum and in the postprandial state but not during the unfed period. When stressed with an HFD, myonectin-knockout male mice had significantly elevated VLDL-triglyceride (TG) and strikingly impaired lipid clearance from circulation following an oral lipid load. Fat distribution between adipose and liver was also altered in myonectin-deficient male mice fed an HFD. Greater fat storage resulted in significantly enlarged adipocytes and was associated with increased postprandial lipoprotein lipase activity in adipose tissue. Parallel to this was a striking reduction in liver steatosis due to significantly reduced TG accumulation. Liver metabolite profiling revealed additional significant changes in bile acids and 1-carbon metabolism pathways. Combined, our data affirm the physiologic importance of myonectin in regulating local and systemic lipid metabolism.-Little, H. C., Rodriguez, S., Lei, X., Tan, S. Y., Stewart, A. N., Sahagun, A., Sarver, D. C., Wong, G. W. Myonectin deletion promotes adipose fat storage and reduces liver steatosis.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/pathology , Cytokines/genetics , Fatty Liver/genetics , Fatty Liver/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Muscle Proteins/genetics , Adipocytes/metabolism , Adipocytes/pathology , Adiposity/genetics , Animals , Cytokines/metabolism , Diet, High-Fat , Fatty Liver/pathology , Female , Homeostasis/genetics , Insulin/genetics , Insulin/metabolism , Insulin Resistance/genetics , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Triglycerides/genetics , Triglycerides/metabolism
14.
Life Sci ; 227: 201-211, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31002917

ABSTRACT

AIMS: Colorectal cancer syndrome has been one of the greatest concerns in the world. Although several epidemiological studies have shown that hepatic low lipoprotein lipase (LPL) mRNA expression may be associated with dyslipidemia and tumor progression, it is still not known whether the liver plays an essential role in hyperlipidemia of ApcMin/+ mice. MAIN METHODS: We measured the expression of metabolic enzymes that involved fatty acid uptake, de novo lipogenesis (DNL), ß-oxidation and investigated hepatic triglyceride production in the liver of wild-type and ApcMin/+ mice. KEY FINDINGS: We found that hepatic fatty acid uptake and DNL decreased, but there was no significant difference in fatty acid ß-oxidation. Interestingly, the production of hepatic very low-density lipoprotein-triglyceride (VLDL-TG) decreased at 20 weeks of age, but marked steatosis was observed in the livers of the ApcMin/+ mouse. To further explore hypertriglyceridemia, we assessed the function of hepatic glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1 (GPIHBP1) for the first time. GPIHBP1 is governed by the transcription factor octamer-binding transcription factor-1 (Oct-1) which are involved in the nuclear factor-κB (NF-κB) signaling pathway in the liver of ApcMin/+ mice. Importantly, it was also confirmed that sn50 (100 µg/mL, an inhibitor of the NF-κB) reversed the tumor necrosis factor α (TNFα)-induced Oct-1 and GPIHBP1 reduction in HepG2 cells. SIGNIFICANCE: Altogether, these findings highlighted a novel role of GPIHBP1 that might be responsible for hypertriglyceridemia in ApcMin/+ mice. Hypertriglyceridemia in these mice may be associated with their hepatic lipid metabolism development.


Subject(s)
Liver/metabolism , Receptors, Lipoprotein/physiology , Triglycerides/metabolism , Animals , Cachexia/metabolism , Cachexia/physiopathology , Colonic Neoplasms/physiopathology , Fatty Acids/metabolism , Fatty Liver/pathology , Gene Expression Regulation/genetics , Hep G2 Cells , Humans , Hyperlipidemias/genetics , Lipid Metabolism/genetics , Lipids/physiology , Lipogenesis/physiology , Lipolysis/physiology , Lipoproteins, VLDL/genetics , Male , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-1/physiology , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Triglycerides/genetics , Tumor Necrosis Factor-alpha/physiology
15.
Int J Mol Sci ; 19(11)2018 Nov 08.
Article in English | MEDLINE | ID: mdl-30413026

ABSTRACT

Rabbits (Oryctolagus cuniculus) are one of the most widely used animal models for the study of human lipid metabolism and atherosclerosis because they are more sensitive to a cholesterol diet than other experimental animals such as rodents. Currently, two hypercholesterolemic rabbit models are frequently used for atherosclerosis studies. One is a cholesterol-fed wild-type rabbit and the other is the Watanabe heritable hyperlipidemic (WHHL) rabbit, which is genetically deficient in low density lipoprotein (LDL) receptor function. Wild-type rabbits can be easily induced to develop severe hypercholesterolemia with a cholesterol-rich diet due to the marked increase in hepatically and intestinally derived remnant lipoproteins, called ß-very low density lipoproteins (VLDL), which are rich in cholesteryl esters. WHHL rabbits are characterized by elevated plasma LDL levels on a standard chow diet, which resembles human familial hypercholesterolemia. Therefore, both rabbit models develop aortic and coronary atherosclerosis, but the elevated plasma cholesterol levels are caused by completely different mechanisms. In addition, cholesterol-fed rabbits but not WHHL rabbits exhibit different degrees of hepatosteatosis. Recently, we along with others have shown that there are many differentially expressed genes in the atherosclerotic lesions and livers of cholesterol-fed rabbits that are either significantly up- or down-regulated, compared with those in normal rabbits, including genes involved in the regulation of inflammation and lipid metabolism. Therefore, dietary cholesterol plays an important role not only in hypercholesterolemia and atherosclerosis but also in hepatosteatosis. In this review, we make an overview of the recent progress in genomic and transcriptomic analyses of hypercholesterolemic rabbits. These transcriptomic profiling data should provide novel insight into the relationship between hypercholesterolemia and atherosclerosis or hepatic dysfunction caused by dietary cholesterol.


Subject(s)
Cholesterol/genetics , Genome/genetics , Hypercholesterolemia/genetics , Transcriptome/genetics , Animals , Cholesterol/metabolism , Disease Models, Animal , Genomics , Humans , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Lipoproteins, VLDL/genetics , Rabbits , Receptors, LDL/genetics
16.
Curr Opin Lipidol ; 29(3): 186-193, 2018 06.
Article in English | MEDLINE | ID: mdl-29708925

ABSTRACT

PURPOSE OF REVIEW: Type 2 diabetes is associated with a characteristic dyslipidemia that may exacerbate cardiovascular risk. The causes of, and the effects of new antihyperglycemia medications on, this dyslipidemia, are under investigation. In an unexpected reciprocal manner, lowering LDL-cholesterol with statins slightly increases the risk of diabetes. Here we review the latest findings. RECENT FINDINGS: The inverse relationship between LDL-cholesterol and diabetes has now been confirmed by multiple lines of evidence. This includes clinical trials, genetic instruments using aggregate single nucleotide polymorphisms, as well as at least eight individual genes - HMGCR, NPC1L1, HNF4A, GCKR, APOE, PCKS9, TM6SF2, and PNPLA3 - support this inverse association. Genetic and pharmacologic evidence suggest that HDL-cholesterol may also be inversely associated with diabetes risk. Regarding the effects of diabetes on lipoproteins, new evidence suggests that insulin resistance but not diabetes per se may explain impaired secretion and clearance of VLDL-triglycerides. Weight loss, bariatric surgery, and incretin-based therapies all lower triglycerides, whereas SGLT2 inhibitors may slightly increase HDL-cholesterol and LDL-cholesterol. SUMMARY: Diabetes and lipoproteins are highly interregulated. Further research is expected to uncover new mechanisms governing the metabolism of glucose, fat, and cholesterol. This topic has important implications for treating type 2 diabetes and cardiovascular disease.


Subject(s)
Cholesterol, HDL , Cholesterol, LDL , Diabetes Mellitus, Type 2 , Dyslipidemias , Lipoproteins, VLDL , Polymorphism, Single Nucleotide , Triglycerides , Animals , Cholesterol, HDL/genetics , Cholesterol, HDL/metabolism , Cholesterol, LDL/genetics , Cholesterol, LDL/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Dyslipidemias/genetics , Dyslipidemias/metabolism , Dyslipidemias/therapy , Humans , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/metabolism , Triglycerides/genetics , Triglycerides/metabolism
17.
Curr Opin Lipidol ; 29(3): 218-223, 2018 06.
Article in English | MEDLINE | ID: mdl-29547398

ABSTRACT

PURPOSE OF REVIEW: Lysosomal acid lipase (LAL), encoded by the LIPA gene, is an essential lysosomal enzyme that hydrolyzes cholesteryl ester and triglyceride delivered to the lysosome. This review highlights the novel pathophysiological role of LAL, the functional genomic discoveries of LIPA as a risk locus for coronary heart diseases (CHD), and the clinical advance in therapies for LAL deficiency. RECENT FINDINGS: The essential role of LAL in lipid metabolism has been confirmed in human and mice with LAL deficiency. In humans, loss-of-function mutations of LIPA cause rare lysosomal disorders, Wolman disease, and cholesteryl ester storage disease, in which LAL enzyme replacement therapy has shown significant benefits in a phase 3 clinical trial. Recent studies have revealed the role of LAL-mediated lysosomal lipolysis in regulating macrophage M2 polarization, lipid mediator production, VLDL secretion, lysosomal function and autophagy, extracellular degradation of aggregated-LDL, and adipose tissue lipolysis. Genome-wide association studies and functional genomic studies have identified LIPA as a risk locus for CHD, but the causal variants and mechanisms remain to be determined. SUMMARY: Despite years of research, our understanding of LAL is incomplete. Future studies will continue to focus on the key pathophysiological functions of LAL in health and diseases including CHD.


Subject(s)
Adipose Tissue/metabolism , Lipid Metabolism , Lipoproteins, VLDL/metabolism , Lysosomes/metabolism , Sterol Esterase/metabolism , Wolman Disease/metabolism , Adipose Tissue/pathology , Animals , Humans , Lipoproteins, VLDL/genetics , Lysosomes/genetics , Lysosomes/pathology , Mice , Sterol Esterase/genetics , Wolman Disease/genetics , Wolman Disease/pathology , Wolman Disease
18.
J Clin Gastroenterol ; 52(5): 444-451, 2018.
Article in English | MEDLINE | ID: mdl-28362682

ABSTRACT

BACKGROUND: Recent genome-wide association studies have identified 2 genetic polymorphisms in association with nonalcoholic fatty liver disease (NAFLD): patatin-like phospholipase domain containing 3 (PNPLA3) and transmembrane 6 superfamily member 2 (TM6SF2), both of which appear to influence the production of very low density lipoprotein (VLDL). The impact of these genetic variations on lipoprotein metabolism in the setting of nonalcoholic steatohepatitis and liver fibrosis are not fully characterized. MATERIALS AND METHODS: We measured comprehensive lipoprotein profiles by nuclear magnetic resonance among 170 serially recruited patients in an NAFLD registry, and determined their relationships with PNPLA3 and TM6SF2 genotypes. RESULTS: In this cohort, 72% patients had at least 1 allele of either PNPLA3 I148M or TM6SF2 E167K, and 30% carried 2 alleles. In multivariate models adjusting for histologic features of nonalcoholic steatohepatitis and liver fibrosis, PNPLA3 I148M is associated with a decrease in VLDL particle size. Both PNPLA3 I148M and TM6SF2 E167K genotypes were associated with increases in the size of low density lipoprotein (LDL) and high density lipoprotein particles, phenotypes considered atheroprotective. When adjusted for both genotypes, NAFLD activity score, in particular the degree of hepatic steatosis was strongly associated with increases in the size of VLDL particles, the concentration of LDL, especially small LDL particles, and a decrease in the size of high density lipoprotein particles, all of which are linked with a proatherogenic phenotype. CONCLUSIONS: PNPLA3 and TM6SF2 are common genetic variants among NAFLD patients and impact lipoprotein profiles in slightly different ways. The interactions between genotypes, hepatic steatosis, and lipoprotein metabolism shed lights on the pathophysiology of NAFLD, and provide opportunities for personalized treatment in the era of emerging NAFLD therapeutics.


Subject(s)
Lipase/genetics , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Non-alcoholic Fatty Liver Disease/genetics , Adult , Aged , Female , Genetic Predisposition to Disease , Genotype , Humans , Lipoproteins, HDL/genetics , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/metabolism , Liver Cirrhosis/pathology , Magnetic Resonance Spectroscopy , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/physiopathology , Polymorphism, Single Nucleotide , Prospective Studies
19.
IUBMB Life ; 70(1): 41-49, 2018 01.
Article in English | MEDLINE | ID: mdl-29281185

ABSTRACT

Hepatitis C virus (HCV) has infected over 170 million people world-wide. This infection causes severe liver damage that can progress to hepatocellular carcinoma leading to death of the infected patients. Development of a cell culture model system for the study of HCV infection in the recent past has helped the researchers world-wide to understand the biology of this virus. Studies over the past decade have revealed the tricks played by the virus to sustain itself, for as long as 40 years, in the host setup without being eliminated by the immune system. Today we understand that the host organelles and different cellular proteins are affected during HCV infection. This cytoplasmic virus has all the cellular organelles at its disposal to successfully replicate, from ribosomes and intracellular membranous structures to the nucleus. It modulates these organelles at both the structural and the functional levels. The vast knowledge about the viral genome and viral proteins has also helped in the development of drugs against the virus. Despite the achieved success rate to cure the infected patients, we struggle to eliminate the cases of recurrence and the non-responders. Such cases might emerge owing to the property of the viral genome to accumulate mutations during its succeeding replication cycles which favours its survival. The current situation calls an urgent need for alternate therapeutic strategies to counter this major problem of human health. © 2017 IUBMB Life, 70(1):41-49, 2018.


Subject(s)
Carcinoma, Hepatocellular/virology , Hepacivirus/pathogenicity , Hepatitis C, Chronic/virology , Hepatocytes/virology , Immune Evasion , Liver Neoplasms/virology , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/prevention & control , Cell Nucleus/immunology , Cell Nucleus/virology , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/virology , Gene Expression Regulation , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Hepatocytes/immunology , Humans , Lipid Droplets/immunology , Lipid Droplets/virology , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/immunology , Liver Neoplasms/etiology , Liver Neoplasms/immunology , Liver Neoplasms/prevention & control , RNA, Viral/biosynthesis , RNA, Viral/genetics , Ribosomes/immunology , Ribosomes/virology , Signal Transduction , Viral Proteins/genetics , Viral Proteins/immunology , Virus Replication/drug effects
20.
Cell Metab ; 26(2): 394-406.e6, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28768177

ABSTRACT

Inhibiting lipogenesis prevents hepatic steatosis in rodents with insulin resistance. To determine if reducing lipogenesis functions similarly in humans, we developed MK-4074, a liver-specific inhibitor of acetyl-CoA carboxylase (ACC1) and (ACC2), enzymes that produce malonyl-CoA for fatty acid synthesis. MK-4074 administered to subjects with hepatic steatosis for 1 month lowered lipogenesis, increased ketones, and reduced liver triglycerides by 36%. Unexpectedly, MK-4074 increased plasma triglycerides by 200%. To further investigate, mice that lack ACC1 and ACC2 in hepatocytes (ACC dLKO) were generated. Deletion of ACCs decreased polyunsaturated fatty acid (PUFA) concentrations in liver due to reduced malonyl-CoA, which is required for elongation of essential fatty acids. PUFA deficiency induced SREBP-1c, which increased GPAT1 expression and VLDL secretion. PUFA supplementation or siRNA-mediated knockdown of GPAT1 normalized plasma triglycerides. Thus, inhibiting lipogenesis in humans reduced hepatic steatosis, but inhibiting ACC resulted in hypertriglyceridemia due to activation of SREBP-1c and increased VLDL secretion.


Subject(s)
Acetyl-CoA Carboxylase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Fatty Liver/blood , Fatty Liver/drug therapy , Triglycerides/blood , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Fatty Liver/genetics , Fatty Liver/pathology , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Lipoproteins, VLDL/genetics , Lipoproteins, VLDL/metabolism , Mice , Mice, Knockout , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Triglycerides/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...