Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 388
Filter
1.
Hepatology ; 78(5): 1418-1432, 2023 11 01.
Article in English | MEDLINE | ID: mdl-36053190

ABSTRACT

BACKGROUND AND AIMS: The assembly and secretion of VLDL from the liver, a pathway that affects hepatic and plasma lipids, remains incompletely understood. We set out to identify players in the VLDL biogenesis pathway by identifying genes that are co-expressed with the MTTP gene that encodes for microsomal triglyceride transfer protein, key to the lipidation of apolipoprotein B, the core protein of VLDL. Using human and murine transcriptomic data sets, we identified small leucine-rich protein 1 ( SMLR1 ), encoding for small leucine-rich protein 1, a protein of unknown function that is exclusively expressed in liver and small intestine. APPROACH AND RESULTS: To assess the role of SMLR1 in the liver, we used somatic CRISPR/CRISPR-associated protein 9 gene editing to silence murine Smlr1 in hepatocytes ( Smlr1 -LKO). When fed a chow diet, male and female mice show hepatic steatosis, reduced plasma apolipoprotein B and triglycerides, and reduced VLDL secretion without affecting microsomal triglyceride transfer protein activity. Immunofluorescence studies show that SMLR1 is in the endoplasmic reticulum and Cis-Golgi complex. The loss of hepatic SMLR1 in female mice protects against diet-induced hyperlipidemia and atherosclerosis but causes NASH. On a high-fat, high-cholesterol diet, insulin and glucose tolerance tests did not reveal differences in male Smlr1 -LKO mice versus controls. CONCLUSIONS: We propose a role for SMLR1 in the trafficking of VLDL from the endoplasmic reticulum to the Cis-Golgi complex. While this study uncovers SMLR1 as a player in the VLDL assembly, trafficking, and secretion pathway, it also shows that NASH can occur with undisturbed glucose homeostasis and atheroprotection.


Subject(s)
Atherosclerosis , Lipoproteins, VLDL , Non-alcoholic Fatty Liver Disease , Small Leucine-Rich Proteoglycans , Animals , Female , Humans , Male , Mice , Apolipoproteins B/blood , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , Leucine , Lipoproteins, VLDL/biosynthesis , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Small Leucine-Rich Proteoglycans/genetics , Small Leucine-Rich Proteoglycans/metabolism , Triglycerides/blood
2.
J Hepatol ; 75(1): 46-54, 2021 07.
Article in English | MEDLINE | ID: mdl-33684506

ABSTRACT

BACKGROUND & AIMS: Excessive fructose intake is associated with increased de novo lipogenesis, blood triglycerides, and hepatic insulin resistance. We aimed to determine whether fructose elicits specific effects on lipid metabolism independently of excessive caloric intake. METHODS: A total of 94 healthy men were studied in this double-blind, randomized trial. They were assigned to daily consumption of sugar-sweetened beverages (SSBs) containing moderate amounts of fructose, sucrose (fructose-glucose disaccharide) or glucose (80 g/day) in addition to their usual diet or SSB abstinence (control group) for 7 weeks. De novo fatty acid (FA) and triglyceride synthesis, lipolysis and plasma free FA (FFA) oxidation were assessed by tracer methodology. RESULTS: Daily intake of beverages sweetened with free fructose and fructose combined with glucose (sucrose) led to a 2-fold increase in basal hepatic fractional secretion rates (FSR) compared to control (median FSR %/day: sucrose 20.8 (p = 0.0015); fructose 19.7 (p = 0.013); control 9.1). Conversely, the same amounts of glucose did not change FSR (median of FSR %/day 11.0 (n.s.)). Fructose intake did not change basal secretion of newly synthesized VLDL-triglyceride, nor did it alter rates of peripheral lipolysis, nor total FA and plasma FFA oxidation. Total energy intake was similar across groups. CONCLUSIONS: Regular consumption of both fructose- and sucrose-sweetened beverages in moderate doses - associated with stable caloric intake - increases hepatic FA synthesis even in a basal state; this effect is not observed after glucose consumption. These findings provide evidence of an adaptative response to regular fructose exposure in the liver. LAY SUMMARY: This study investigated the metabolic effects of daily sugar-sweetened beverage consumption for several weeks in healthy lean men. It revealed that beverages sweetened with the sugars fructose and sucrose (glucose and fructose combined), but not glucose, increase the ability of the liver to produce lipids. This change may pave the way for further unfavorable effects on metabolic health. CLINICAL TRIAL REGISTRATION NUMBER: NCT01733563.


Subject(s)
Fatty Acids/biosynthesis , Fructose , Glucose , Lipogenesis , Lipoproteins, VLDL/biosynthesis , Liver , Sucrose , Triglycerides/biosynthesis , Adult , Double-Blind Method , Energy Intake , Fructose/administration & dosage , Fructose/adverse effects , Fructose/metabolism , Glucose/administration & dosage , Glucose/metabolism , Healthy Volunteers , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipogenesis/drug effects , Lipogenesis/physiology , Liver/drug effects , Liver/metabolism , Male , Sucrose/administration & dosage , Sucrose/adverse effects , Sucrose/metabolism , Sugar-Sweetened Beverages , Sweetening Agents/pharmacology
3.
Cell Rep ; 34(11): 108873, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33730569

ABSTRACT

Contacts between organelles create microdomains that play major roles in regulating key intracellular activities and signaling pathways, but whether they also regulate systemic functions remains unknown. Here, we report the ultrastructural organization and dynamics of the inter-organellar contact established by sheets of curved rough endoplasmic reticulum closely wrapped around the mitochondria (wrappER). To elucidate the in vivo function of this contact, mouse liver fractions enriched in wrappER-associated mitochondria are analyzed by transcriptomics, proteomics, and lipidomics. The biochemical signature of the wrappER points to a role in the biogenesis of very-low-density lipoproteins (VLDL). Altering wrappER-mitochondria contacts curtails VLDL secretion and increases hepatic fatty acids, lipid droplets, and neutral lipid content. Conversely, acute liver-specific ablation of Mttp, the most upstream regulator of VLDL biogenesis, recapitulates this hepatic dyslipidemia phenotype and promotes remodeling of the wrappER-mitochondria contact. The discovery that liver wrappER-mitochondria contacts participate in VLDL biology suggests an involvement of inter-organelle contacts in systemic lipid homeostasis.


Subject(s)
Endoplasmic Reticulum/metabolism , Homeostasis , Lipids/chemistry , Liver/metabolism , Mitochondria/metabolism , Animals , Endoplasmic Reticulum/ultrastructure , Enterocytes/metabolism , Gene Silencing , Hepatocytes/metabolism , Imaging, Three-Dimensional , Intestine, Small/cytology , Lipoproteins, VLDL/biosynthesis , Male , Metabolomics , Mice, Inbred C57BL , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Phospholipids/biosynthesis , Proteins/metabolism
4.
Nutrients ; 12(6)2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32549314

ABSTRACT

A high fructose intake exacerbates postprandial plasma triacylglycerol (TAG) concentration, an independent risk factor for cardiovascular disease, although it is unclear whether this is due to increased production or impaired clearance of triacylglycerol (TAG)-rich lipoproteins. We determined the in vivo acute effect of fructose on postprandial intestinal and hepatic lipoprotein TAG kinetics and de novo lipogenesis (DNL). Five overweight men were studied twice, 4 weeks apart. They consumed hourly mixed-nutrient drinks that were high-fructose (30% energy) or low-fructose (<2% energy) for 11 h. Oral 2H2O was administered to measure fasting and postprandial DNL. Postprandial chylomicron (CM)-TAG and very low-density lipoprotein (VLDL)-TAG kinetics were measured with an intravenous bolus of [2H5]-glycerol. CM and VLDL were separated by their apolipoprotein B content using antibodies. Plasma TAG (p < 0.005) and VLDL-TAG (p = 0.003) were greater, and CM-TAG production rate (PR, p = 0.046) and CM-TAG fractional catabolic rate (FCR, p = 0.073) lower when high-fructose was consumed, with no differences in VLDL-TAG kinetics. Insulin was lower (p = 0.005) and apoB48 (p = 0.039), apoB100 (p = 0.013) and non-esterified fatty acids (NEFA) (p = 0.013) were higher after high-fructose. Postprandial hepatic fractional DNL was higher than intestinal fractional DNL with high-fructose (p = 0.043) and low-fructose (p = 0.043). Fructose consumption had no effect on the rate of intestinal or hepatic DNL. We provide the first measurement of the rate of intestinal DNL in humans. Lower CM-TAG PR and CM-TAG FCR with high-fructose consumption suggests lower clearance of CM, rather than elevated production, may contribute to elevated plasma TAG, possibly due to lower insulin-mediated stimulation of lipoprotein lipase.


Subject(s)
Fatty Acids/biosynthesis , Fructose/administration & dosage , Intestines/drug effects , Lipogenesis/drug effects , Triglycerides/biosynthesis , Adult , Beverages , Chylomicrons/biosynthesis , Diet , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipoproteins, VLDL/biosynthesis , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Postprandial Period/physiology , Triglycerides/blood
5.
Am J Physiol Endocrinol Metab ; 318(2): E262-E275, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31821038

ABSTRACT

miR-130b is a microRNA whose expression is particularly elevated within adipose tissue and in the circulation in diabetic states. Hepatic miR-130b expression has been linked to hepatocellular carcinoma and changes in lipid metabolism. Here, we investigated the role of miR-130b in hepatic lipid homeostasis and lipoprotein export. We observed that overexpression of miR-130b-3p or -5p in HepG2 cells markedly enhanced the secretion of very-low-density lipoprotein (VLDL) particles, enhanced the secretion of [3H]glycerol metabolically labeled triglyceride (TG), and significantly increased the number or the average size of lipid droplets (LDs), respectively. Overexpression of miR-130b also altered the expression of key genes involved in lipid metabolism and in particular markedly increased both mRNA and protein expression levels of microsomal triglyceride transfer protein (MTP). Conversely, the miR-130b inhibitor decreased mRNA levels of MTP and fatty acid synthase (FAS) in HepG2 cells. However, dual-luciferase reporter assays indicated that MTP is not a direct target of miR-130b-3p. miR-130b overexpression did not alter de novo synthesized TG or the stability and secretion of apolipoprotein B 100. Interestingly, knockdown of phosphatase and tensin homolog (PTEN) blocked the upregulation of MTP mRNA induced by miR-130b. Finally, miR-130b-induced stimulation of VLDL secretion was also observed in a second hepatocyte cell culture model, immortalized human hepatocytes, confirming the effects observed in HepG2 cells. Overall, these data suggest a potential role for miR-130b in promoting hepatic VLDL assembly and secretion mediated by marked stimulation of MTP expression and TG mobilization. Thus miR-130b overexpression corrects the defect in VLDL production in HepG2 cells.


Subject(s)
Carrier Proteins/biosynthesis , Lipoproteins, VLDL/biosynthesis , Lipoproteins, VLDL/metabolism , Liver/metabolism , MicroRNAs/metabolism , Microsomes, Liver/enzymology , Apolipoprotein B-100/biosynthesis , Apolipoprotein B-100/genetics , Cell Line , Cells, Cultured , Fatty Acid Synthesis Inhibitors/pharmacology , Gene Knockdown Techniques , Hepatocytes/metabolism , Humans , Lipid Metabolism/genetics , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism
6.
Diabetes Metab Syndr ; 13(2): 1529-1534, 2019.
Article in English | MEDLINE | ID: mdl-31336517

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) is an integral cell-membrane protein that mediates the rate-limiting step of high density lipoprotein (HDL) biogenesis and suppression of inflammation by triggering a number of signaling pathways via interacting with an apolipoprotein acceptor. The hepatic ABCA1 is involved in regulation of very low density lipoprotein (VLDL) production by affecting the apolipoprotein B trafficking and lipidation of VLDL particles. This protein is involved in protecting the function of pancreatic ß-cells and insulin secretion by cholesterol homeostasis. Adipose tissue lipolysis is associated with ABCA1 activity. This transporter is involved in controlling obesity and insulin sensitivity by regulating triglyceride (TG) lipolysis and influencing on adiponectin, visfatin, leptin, and GLUT4 genes expression. The ABCA1 of skeletal muscle cells play a role in increasing the glucose uptake by enhancing the Akt phosphorylation and transferring GLUT4 to the plasma membrane. Abnormal status of ABCA1-regulated phenotypes is observed in metabolic syndrome. This syndrome is associated with the occurrence of many diseases. This review is a summary of the role of ABCA1 in HDL and VLDL production, homeostasis of insulin and glucose, suppression of inflammation and obesity controlling to provide a better insight into the association of this protein with metabolic syndrome.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Glucose/metabolism , Inflammation/physiopathology , Insulin/metabolism , Lipoproteins, HDL/biosynthesis , Lipoproteins, VLDL/biosynthesis , Metabolic Syndrome/epidemiology , Obesity/physiopathology , Homeostasis , Humans , Iran/epidemiology
7.
Can J Physiol Pharmacol ; 97(11): 1018-1027, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31247150

ABSTRACT

ARHGAP21 is a Rho-GAP that controls GTPases activity in several tissues, but its role on liver lipid metabolism is unknown. Thus, to achieve the Rho-GAP role in the liver, control and ARHGAP21-haplodeficient mice were fed chow (Ctl and Het) or high-fat diet (Ctl-HFD and Het-HFD) for 12 weeks, and pyruvate and insulin tolerance tests, insulin signaling, liver glycogen and triglycerides content, gene and protein expression, and very-low-density lipoprotein secretion were measured. Het mice displayed reduced body weight and plasma triglycerides levels, and increased liver insulin signaling. Reduced gluconeogenesis and increased glycogen content were observed in Het-HFD mice. Gene and protein expression of microsomal triglyceride transfer protein were reduced in both Het mice, while the lipogenic genes SREBP-1c and ACC were increased. ARHGAP21 knockdown resulted in hepatic steatosis through increased hepatic lipogenesis activity coupled with decreases in CPT1a expression and very-low-density lipoprotein export. In conclusion, liver of ARHGAP21-haplodeficient mice are more insulin sensitive, associated with higher lipid synthesis and lower lipid export.


Subject(s)
GTPase-Activating Proteins/deficiency , Gene Knockout Techniques , Insulin/metabolism , Lipid Metabolism , Liver/metabolism , Obesity/metabolism , Obesity/pathology , Animals , GTPase-Activating Proteins/genetics , Glucose/biosynthesis , Glycogen/metabolism , Lipid Metabolism/genetics , Lipoproteins, VLDL/biosynthesis , Lipoproteins, VLDL/metabolism , Liver/pathology , Mice , Signal Transduction/genetics
8.
Commun Biol ; 2: 173, 2019.
Article in English | MEDLINE | ID: mdl-31098406

ABSTRACT

Medicinal plants show important therapeutic value in chronic disease treatment. However, due to their diverse ingredients and complex biological effects, the molecular mechanisms of medicinal plants are yet to be explored. By means of several high-throughput platforms, here we show hawk tea extract (HTE) inhibits Niemann-Pick C1-like 1 (NPC1L1)-mediated free cholesterol uptake, thereby inducing the transcription of low-density lipoprotein receptor (LDLR) downstream of the sterol response element binding protein 2 (SREBP2) pathway. Meanwhile, HTE suppresses hepatocyte nuclear factor 4α (HNF4α)-mediated transcription of microsomal triglyceride transfer protein (MTP) and apolipoprotein B (APOB), thereby decreasing the production of very-low-density lipoprotein. The catechin EGCG ((-)-epigallocatechin gallate) and the flavonoids kaempferol and quercetin are identified as the bioactive components responsible for the effects on the NPC1L1-SREBP2-LDLR axis and HNF4α-MTP/APOB axis, respectively. Overall, hawk tea works as a previously unrecognized cholesterol-lowering agent in a multi-target and multi-component manner.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol/metabolism , Lipoproteins, VLDL/biosynthesis , Litsea , Teas, Medicinal , Animals , Anticholesteremic Agents/chemistry , Biological Transport, Active/drug effects , Caffeine/analysis , Catechin/analogs & derivatives , Catechin/pharmacology , Disease Models, Animal , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Feeder Cells , Gastrointestinal Microbiome/drug effects , Hep G2 Cells , Humans , Kaempferols/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Litsea/chemistry , Liver/drug effects , Liver/metabolism , Male , Models, Biological , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, LDL/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Teas, Medicinal/analysis
9.
Arterioscler Thromb Vasc Biol ; 39(3): 373-386, 2019 03.
Article in English | MEDLINE | ID: mdl-30700132

ABSTRACT

Objective- APOA5 variants are strongly associated with hypertriglyceridemia, as well as increased risks of cardiovascular disease and acute pancreatitis. Hypertriglyceridemia in apo AV dysfunction often aggravates by environmental factors such as high-carbohydrate diets or aging. To date, the molecular mechanisms by which these environmental factors induce hypertriglyceridemia are poorly defined, leaving the high-risk hypertriglyceridemia condition undertreated. Previously, we reported that LXR (liver X receptor)-SREBP (sterol regulatory element-binding protein)-1c pathway regulates large-VLDL (very low-density lipoprotein) production induced by LXR agonist. However, the pathophysiological relevance of the finding remains unknown. Approach and Results- Here, we reconstitute the environment-induced hypertriglyceridemia phenotype of human APOA5 deficiency in Apoa5-/- mice and delineate the role of SREBP-1c in vivo by generating Apoa5-/- ;Srebp-1c-/- mice. The Apoa5-/- mice, which showed moderate hypertriglyceridemia on a chow diet, developed severe hypertriglyceridemia on high-carbohydrate feeding or aging as seen in patients with human apo AV deficiency. These responses were nearly completely abolished in the Apoa5-/- ;Srebp-1c-/- mice. Further mechanistic studies revealed that in response to these environmental factors, SREBP-1c was activated to increase triglyceride synthesis and to permit the incorporation of triglyceride into abnormally large-VLDL particles, which require apo AV for efficient clearance. Conclusions- Severe hypertriglyceridemia develops only when genetic factors (apo AV deficiency) and environmental effects (SREBP-1c activation) coexist. We demonstrate that the regulated production of large-sized VLDL particles via SREBP-1c determines plasma triglyceride levels in apo AV deficiency. Our findings explain the long-standing enigma of the late-onset hypertriglyceridemia phenotype of apo AV deficiency and suggest a new approach to treat hypertriglyceridemia by targeting genes that mediate environmental effects.


Subject(s)
Apolipoprotein A-V/deficiency , Hypertriglyceridemia/blood , Lipoproteins, VLDL/biosynthesis , Sterol Regulatory Element Binding Protein 1/physiology , Aging/metabolism , Animal Feed/adverse effects , Animals , Apolipoprotein A-V/genetics , Apolipoproteins/blood , Chylomicrons/metabolism , Female , Fructose/toxicity , Gene Expression Regulation/drug effects , Gene-Environment Interaction , Humans , Hydrocarbons, Fluorinated/pharmacology , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/genetics , Lipids/blood , Liver X Receptors/agonists , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Olive Oil/toxicity , Sterol Regulatory Element Binding Protein 1/deficiency , Sterol Regulatory Element Binding Protein 1/genetics , Sulfonamides/pharmacology
10.
Obes Res Clin Pract ; 12(Suppl 2): 16-28, 2018.
Article in English | MEDLINE | ID: mdl-27133527

ABSTRACT

BACKGROUND: 5-HT has been shown to mediate abnormality of hepatic lipid metabolism through activation of mammalian target of rapamycin (mTOR). However, it is unclear whether 5-HT is directly involved in high-fat diet (HFD)-induced hepatic steatosis. MATERIALS AND METHODS: Male rats were allocated into seven groups with control, either HFD feeding, 5-HT treatment, or HFD feeding and 5-HT treatment with or without sarpogrelate treatment, all of which were executed for 4 weeks. HepG2 cells were exposed to 5-HT or palmitic acid (PA) with or without rapamycin or Sar treatment. RESULTS: Rats fed with HFD or exposed to 5-HT led to abnormalities with activated hepatic mTOR-S6K pathway, overproduction of hepatic triglycerides and VLDL with steatosis, and hyperlipidemia, which were exacerbated by a combination of HFD and 5-HT. Sarpogrelate significantly inhibited above abnormalities induced by HFD and 5-HT, alone or in a combination. Additionally, HFD caused up-regulation of 5-HT2 receptors (5-HT2R), including 5-HT2AR and 5-HT2BR, and 5-HT synthesis in the liver, without obvious influence on other 5-HT receptors gene expression. In HepG2 cells, both PA and 5-HT induced overproduction of triglycerides and VLDL with lipid droplets, and PA up-regulated 5-HT2AR and 5-HT2BR expression and 5-HT synthesis as well. Rapamycin fully abolished PA or 5-HT-induced mTOR activation, which was more effective than sarpogrelate. However, the inhibitory effects of rapamycin on PA or 5-HT-induced overproduction of triglycerides and VLDL were less than sarpogrelate. CONCLUSIONS: Up-regulation of hepatic 5-HT2R and 5-HT synthesis by HFD is crucial for HFD-induced overproduction of hepatic triglycerides and VLDL with hyperlipidemia.


Subject(s)
Cholesterol, VLDL/biosynthesis , Diet, High-Fat , Fatty Liver/metabolism , Lipoproteins, VLDL/biosynthesis , Receptors, Serotonin, 5-HT2/metabolism , Animals , Cholesterol, VLDL/blood , Disease Models, Animal , Fatty Liver/blood , Lipoproteins, VLDL/blood , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley , Up-Regulation
11.
Mol Nutr Food Res ; 62(5)2018 03.
Article in English | MEDLINE | ID: mdl-29210166

ABSTRACT

SCOPE: This study aims to characterize the effect of fenugreek (Trigonella foenum-graecum) seed and its phytoceutical trigonelline in antimetabolic inflammation and ameliorating overproduction of very low density lipoprotein (VLDL) in insulin resistance. METHODS AND RESULTS: Two groups of genetic hyperlipidemic mice generated by depletion of cAMP responsive element binding protein H (CREBH) are fed either a chow containing 2% fenugreek seed or vehicle for 7 weeks. Q-RT-PCR and immunoblotting analysis demonstrated that fenugreek seed containing diet inhibits hepatic SREBP-1c activation and the subsequent de novo lipogenesis by enhancing expression of insulin-inducible gene-1 (Insig-1) and gene-2 (Insig-2). mRNA expression of PPARα and its target genes that are involved in fatty acid ß-oxidation are also upregulated in the fenugreek seed fed-mice which is accompanied by significantly reduced hepatic lipid accumulation and VLDL secretion, improved endoplasmic reticulum (ER) stress, and ameliorated metabolic inflammation. These actions enhance insulin sensitivity and improve hyperlipidemia. In vitro, treating a rat hepatoma cell line, McA-RH7777 (McA), with trigonelline is able to recapitulate the results observed in vivo. CONCLUSIONS: This study unveils a novel mechanism of fenugreek seed and trigonelline in countering hepatic VLDL overproduction and insulin resistance by enhancing the Insig signaling pathways and ameliorating metabolic inflammatory stress in the liver.


Subject(s)
Alkaloids/pharmacology , Insulin Resistance , Lipoproteins, VLDL/antagonists & inhibitors , Membrane Proteins/physiology , Plant Extracts/pharmacology , Signal Transduction/drug effects , Animals , Endoplasmic Reticulum Stress/drug effects , Lipogenesis/drug effects , Lipoproteins, VLDL/biosynthesis , Mice , Rats , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 1/physiology , Trigonella , Tumor Cells, Cultured
12.
Environ Toxicol Pharmacol ; 56: 15-20, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28866046

ABSTRACT

Formaldehyde is a common indoor air pollutant that is toxic to the liver. This study aimed to investigate the effects of formaldehyde on triglyceride metabolism in human hepatocellular carcinoma cells (HepG2). Cell viability was detected using a MTT (3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide) assay. Following treatment with different concentrations of formaldehyde for 24 and 48h, the intra and extra-hepatocellular triglyceride (TG) content was determined using a chemical-enzymatic method; Western blotting was used to detect the levels of fatty acid synthesis and VLDL-related proteins. Our results showed that cell viability significantly decreased after formaldehyde treatment (0.5-12.5mM, 24/48h). Extracellular TG levels in the hepatocytes increased after formaldehyde treatment at 0.004mM-0.1mM for 24h. SREBP-1c, ACC, FASN, and MTP, CES3 and DGAT1 proteins increased significantly after 24h of formaldehyde treatment. Intracellular TG levels decreased for 48h treatment of formaldehyde. AMPKα increased significantly in all tested groups and p-AMPK increased significantly after 0.1mM formaldehyde treatment for 48h. Our results indicated that short-term formaldehyde exposure balances triglyceride metabolism by promoting hepatocellular TG synthesis and VLDL secretion; Long-term formaldehyde disturbs the TG metabolism balance in the hepatocytes.


Subject(s)
Formaldehyde/pharmacology , Hepatocytes/cytology , Lipoproteins, VLDL/biosynthesis , Triglycerides/biosynthesis , AMP-Activated Protein Kinases/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Time Factors
13.
J Dairy Res ; 83(4): 442-446, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27692001

ABSTRACT

Dairy cows with fatty liver or ketosis exhibit hyperketonemia, oxidative stress, and a low rate of very low density lipoprotein (VLDL) assembly, and there may be a potential link among these characteristics. Therefore, the objective of this study was to determine the effect of acetoacetic acid (AcAc) on the assembly of VLDL in cow hepatocytes. Cultured cow hepatocytes were treated with different concentrations of AcAc with or without N-acetylcysteine (NAC, an antioxidant). AcAc treatment decreased the mRNA expression and activities of antioxidant enzymes superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px), and significantly increased malondialdehyde (MDA) content, indicative of oxidative stress. Furthermore, AcAc treatment significantly down-regulated the mRNA expression of apolipoprotein B100 (ApoB100), apolipoprotein E (ApoE), and low density lipoprotein receptor (LDLR), which thus decreased VLDL assembly and increased triglyceride (TG) accumulation in these bovine hepatocytes. Importantly, NAC relieved AcAc-induced oxidative stress and increased VLDL assembly. In summary, these results suggest that AcAc-induced oxidative stress affects the assembly of VLDL, which increases TG accumulation in bovine hepatocytes.


Subject(s)
Acetoacetates/pharmacology , Cattle , Hepatocytes/metabolism , Lipoproteins, VLDL/biosynthesis , Oxidative Stress/drug effects , Acetoacetates/administration & dosage , Animals , Antioxidants/administration & dosage , Antioxidants/analysis , Cells, Cultured , China , Female , Gene Expression/drug effects , Hepatocytes/chemistry , Hepatocytes/drug effects , RNA, Messenger/analysis , Triglycerides/metabolism
14.
Nat Rev Mol Cell Biol ; 16(11): 678-89, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26490400

ABSTRACT

Fatty acid and fat synthesis in the liver is a highly regulated metabolic pathway that is important for very low-density lipoprotein (VLDL) production and thus energy distribution to other tissues. Having common features at their promoter regions, lipogenic genes are coordinately regulated at the transcriptional level. Transcription factors, such as upstream stimulatory factors (USFs), sterol regulatory element-binding protein 1C (SREBP1C), liver X receptors (LXRs) and carbohydrate-responsive element-binding protein (ChREBP) have crucial roles in this process. Recently, insights have been gained into the signalling pathways that regulate these transcription factors. After feeding, high blood glucose and insulin levels activate lipogenic genes through several pathways, including the DNA-dependent protein kinase (DNA-PK), atypical protein kinase C (aPKC) and AKT-mTOR pathways. These pathways control the post-translational modifications of transcription factors and co-regulators, such as phosphorylation, acetylation or ubiquitylation, that affect their function, stability and/or localization. Dysregulation of lipogenesis can contribute to hepatosteatosis, which is associated with obesity and insulin resistance.


Subject(s)
Fatty Acids/biosynthesis , Lipogenesis/genetics , Lipoproteins, VLDL/biosynthesis , Liver/metabolism , Transcription, Genetic/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , DNA-Activated Protein Kinase/metabolism , Gene Expression Regulation , Lipogenesis/physiology , Liver X Receptors , Mice , Nuclear Proteins/metabolism , Orphan Nuclear Receptors/metabolism , Protein Kinase C/metabolism , Protein Processing, Post-Translational/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Upstream Stimulatory Factors/metabolism
15.
J Clin Endocrinol Metab ; 100(11): 4098-105, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26291068

ABSTRACT

CONTEXT: Subjects with type 2 diabetes mellitus (T2DM) and diabetic nephropathy (DN) often exhibit hypertriglyceridemia. The mechanism(s) of such an increase are poorly known. OBJECTIVE: We investigated very low-density lipoprotein (VLDL)-Apo B 100 kinetics in T2DM subjects with and without DN, and in healthy controls. DESIGN: Stable isotope (13)C-leucine infusion and modeling analysis of tracer-to-tracee ratio dynamics in the protein product pool in the 6-8-h period following tracer infusion were employed. SETTING: Male subjects affected by T2DM, either with (n = 9) or without (n = 5) DN, and healthy male controls (n = 6), were studied under spontaneous glycemic levels in the post-absorptive state. RESULTS: In the T2DM patients with DN, plasma triglyceride (TG) (mean ± SD; 2.2 ± 0.8 mmol/L) and VLDL-Apo B 100 (17.4 ± 10.4 mg/dL) concentrations, and VLDL-Apo B 100 pool (0.56 ± 0.29 g), were ∼60-80% greater (P < .05 or less) than those of the T2DM subjects without DN (TG, 1.4 ± 0.5 mmol/L; VLDL-Apo B 100, 9.9 ± 2.5 mg/dL; VLDL-Apo B 100 pool, 0.36 ± 0.09 g), and ∼80-110% greater (P < .04 or less) than those of nondiabetic controls (TG, 1.2 ± 0.4 mmol/L; VLDL-Apo B 100, 8.2 ± 1.7 mg/dL; VLDL-Apo B 100, 0.32 ± 0.09 g). In sharp contrast however, in the subjects with T2DM and DN, VLDL-Apo B 100 fractional synthesis rate was ≥50% lower (4.8 ± 2.2 pools/d) than that of either the T2DM subjects without DN (9.9 ± 4.3 pools/d; P < .025) or the control subjects (12.5 ± 9.1 pools/d; P < .04). CONCLUSIONS: The hypertriglyceridemia of T2DM patients with DN is not due to hepatic VLDL-Apo B 100 overproduction, which is decreased, but it should be attributed to decreased apolipoprotein removal.


Subject(s)
Apolipoprotein B-100/biosynthesis , Diabetes Mellitus, Type 2/blood , Diabetic Nephropathies/blood , Hypertriglyceridemia/blood , Lipoproteins, VLDL/biosynthesis , Adult , Aged , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/diagnostic imaging , Diabetic Nephropathies/diagnostic imaging , Female , Humans , Insulin/blood , Kinetics , Leucine , Male , Middle Aged , Radionuclide Imaging , Radiopharmaceuticals
16.
J Lipid Res ; 56(7): 1282-95, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25964513

ABSTRACT

LCAT, a plasma enzyme that esterifies cholesterol, has been proposed to play an antiatherogenic role, but animal and epidemiologic studies have yielded conflicting results. To gain insight into LCAT and the role of free cholesterol (FC) in atherosclerosis, we examined the effect of LCAT over- and underexpression in diet-induced atherosclerosis in scavenger receptor class B member I-deficient [Scarab(-/-)] mice, which have a secondary defect in cholesterol esterification. Scarab(-/-)×LCAT-null [Lcat(-/-)] mice had a decrease in HDL-cholesterol and a high plasma ratio of FC/total cholesterol (TC) (0.88 ± 0.033) and a marked increase in VLDL-cholesterol (VLDL-C) on a high-fat diet. Scarab(-/-)×LCAT-transgenic (Tg) mice had lower levels of VLDL-C and a normal plasma FC/TC ratio (0.28 ± 0.005). Plasma from Scarab(-/-)×LCAT-Tg mice also showed an increase in cholesterol esterification during in vitro cholesterol efflux, but increased esterification did not appear to affect the overall rate of cholesterol efflux or hepatic uptake of cholesterol. Scarab(-/-)×LCAT-Tg mice also displayed a 51% decrease in aortic sinus atherosclerosis compared with Scarab(-/-) mice (P < 0.05). In summary, we demonstrate that increased cholesterol esterification by LCAT is atheroprotective, most likely through its ability to increase HDL levels and decrease pro-atherogenic apoB-containing lipoprotein particles.


Subject(s)
Atherosclerosis/blood , Atherosclerosis/enzymology , CD36 Antigens/deficiency , CD36 Antigens/genetics , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Biological Transport , Blood Platelets/metabolism , Blood Platelets/pathology , Cholesterol/blood , Erythrocyte Count , Erythrocytes/metabolism , Erythrocytes/pathology , Esterification , Female , Gene Expression Regulation, Enzymologic , Gene Knockout Techniques , Humans , Lipoproteins, VLDL/biosynthesis , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/chemistry , Liver/metabolism , Mice , Mice, Transgenic , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Platelet Count
17.
Br J Nutr ; 112(10): 1592-600, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25263431

ABSTRACT

The aim of the present study was to identify molecular mechanisms involved in liver fat and cholesterol accumulation in ovariectomised (Ovx) rats fed with high-cholesterol diets. VLDL assembly and bile acid metabolism were specifically targeted. After being either Ovx or sham-operated, the rats were fed a standard diet or a high-fat diet containing 0, 0·25 or 0·5 % cholesterol for 6 weeks. Although Ovx rats exposed to dietary cholesterol intake accumulated the greatest amount of hepatic fat and cholesterol, plasma cholesterol levels were lower (P< 0·05) in these animals than in the corresponding control rats. Accompanying this observation, ovariectomy and dietary cholesterol intake resulted in a down-regulation (P< 0·05) of the expression of genes associated with VLDL assembly, including microsomal TAG transfer protein, diacylglycerol acyltransferase 2, acyl-CoA:cholesterol acyltransferase 2 and apoB-100 as well as genes associated with bile acid metabolism including farnesoid X receptor and bile salt export pump (P< 0·01). These results indicate that high-fat/high-cholesterol diets and ovariectomy concomitantly disrupt hepatic lipid output through defects in VLDL assembly and, most probably, secretion. The results also point to a defect in hepatic bile acid secretion. The present study offers novel insights into intrahepatic lipid metabolism, which may be relevant to metabolic complications found in postmenopausal women.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Fats/metabolism , Estrogens/deficiency , Fatty Liver/metabolism , Lipid Metabolism , Lipoproteins, VLDL/biosynthesis , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein B-100/metabolism , Bile Acids and Salts/metabolism , Carrier Proteins/metabolism , Cholesterol, Dietary/adverse effects , Cholesterol, Dietary/metabolism , Dietary Fats/adverse effects , Down-Regulation , Fatty Liver/etiology , Female , Lipoproteins, VLDL/metabolism , Ovariectomy , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Sterol O-Acyltransferase/metabolism , Sterol O-Acyltransferase 2
18.
J Dairy Sci ; 97(3): 1328-35, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24377799

ABSTRACT

High serum concentrations of nonesterified fatty acids (NEFA), which may affect the synthesis and assembly of very low density lipoproteins (VLDL), are associated with fatty liver during the early lactation period. Therefore, the objective of this study was to investigate the effects of NEFA on the synthesis and assembly of VLDL in bovine hepatocytes. Bovine hepatocytes were cultured and treated with different concentrations of NEFA. The mRNA expression of apolipoprotein B100 (ApoB100) and apolipoprotein E (ApoE) was significantly lower in the NEFA treatment groups than in the control group (0mM NEFA). The abundance of mRNA from microsomal triglyceride transfer protein (MTP) and the low density lipoprotein receptor (LDLR) was significantly lower in the medium- and high-dose NEFA treatment groups. The protein expression of ApoB100, ApoE, MTP, and LDLR was found to be significantly and dose-dependently decreased in groups of NEFA-treated hepatocytes. The VLDL content was also significantly decreased in the NEFA-treated hepatocytes. Large amounts of triglycerides accumulated in the hepatocytes. These results indicate that NEFA significantly inhibits the expression of ApoB100, ApoE, MTP, and LDLR, thereby decreasing the synthesis and assembly of VLDL and inducing TG accumulation in bovine hepatocytes.


Subject(s)
Fatty Acids, Nonesterified/blood , Hepatocytes/drug effects , Lipoproteins, VLDL/biosynthesis , Animals , Apolipoprotein B-100/genetics , Apolipoprotein B-100/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cattle , Cells, Cultured , Hepatocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Triglycerides/metabolism
19.
Clin Chem Lab Med ; 52(12): 1695-727, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23940067

ABSTRACT

Research into lipoprotein metabolism has developed because understanding lipoprotein metabolism has important clinical indications. Lipoproteins are risk factors for cardiovascular disease. Recent advances include the identification of factors in the synthesis and secretion of triglyceride rich lipoproteins, chylomicrons (CM) and very low density lipoproteins (VLDL). These included the identification of microsomal transfer protein, the cotranslational targeting of apoproteinB (apoB) for degradation regulated by the availability of lipids, and the characterization of transport vesicles transporting primordial apoB containing particles to the Golgi. The lipase maturation factor 1, glycosylphosphatidylinositol-anchored high density lipoprotein binding protein 1 and an angiopoietin-like protein play a role in lipoprotein lipase (LPL)-mediated hydrolysis of secreted CMs and VLDL so that the right amount of fatty acid is delivered to the right tissue at the right time. Expression of the low density lipoprotein (LDL) receptor is regulated at both transcriptional and post-transcriptional level. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has a pivotal role in the degradation of LDL receptor. Plasma remnant lipoproteins bind to specific receptors in the liver, the LDL receptor, VLDL receptor and LDL receptor-like proteins prior to removal from the plasma. Reverse cholesterol transport occurs when lipid free apoAI recruits cholesterol and phospholipid to assemble high density lipoprotein (HDL) particles. The discovery of ABC transporters (ABCA1 and ABCG1) and scavenger receptor class B type I (SR-BI) provided further information on the biogenesis of HDL. In humans HDL-cholesterol can be returned to the liver either by direct uptake by SR-BI or through cholesteryl ester transfer protein exchange of cholesteryl ester for triglycerides in apoB lipoproteins, followed by hepatic uptake of apoB containing particles. Cholesterol content in cells is regulated by several transcription factors, including the liver X receptor and sterol regulatory element binding protein. This review summarizes recent advances in knowledge of the molecular mechanisms regulating lipoprotein metabolism.


Subject(s)
Lipoproteins/metabolism , ATP-Binding Cassette Transporters/metabolism , Apolipoproteins/biosynthesis , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cholesterol Ester Transfer Proteins/metabolism , Chylomicrons/metabolism , Humans , Lipoproteins, HDL/biosynthesis , Lipoproteins, VLDL/biosynthesis , Phospholipid Transfer Proteins/metabolism , Receptors, Scavenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...