Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Diabetes ; 72(2): 210-222, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36346641

ABSTRACT

Cellular lipid storage is regulated by the balance of lipogenesis and lipolysis. The rate-limiting triglyceride hydrolase ATGL (desnutrin/PNPLA2) is critical for lipolysis. The control of ATGL transcription, localization, and activation has been intensively studied, while regulation of the protein stability of ATGL is much less explored. In this study, we showed that the protein stability of ATGL is regulated by the N-end rule in cultured cells and in mice. The N-end rule E3 ligases UBR1 and UBR2 reduce the level of ATGL and affect lipid storage. The N-end rule-resistant ATGL(F2A) mutant, in which the N-terminal phenylalanine (F) of ATGL is substituted by alanine (A), has increased protein stability and enhanced lipolysis activity. ATGLF2A/F2A knock-in mice are protected against high-fat diet (HFD)-induced obesity, hepatic steatosis, and insulin resistance. Hepatic knockdown of Ubr1 attenuates HFD-induced hepatic steatosis by enhancing the ATGL level. Finally, the protein levels of UBR1 and ATGL are negatively correlated in the adipose tissue of obese mice. Our study reveals N-end rule-mediated proteasomal regulation of ATGL, a finding that may potentially be beneficial for treatment of obesity.


Subject(s)
Acyltransferases , Fatty Liver , Lipase , Animals , Mice , Adipose Tissue/metabolism , Fatty Liver/genetics , Fatty Liver/metabolism , Lipase/genetics , Lipase/metabolism , Lipolysis/genetics , Obesity/metabolism , Triglycerides/metabolism , Acyltransferases/metabolism , Proteasome Endopeptidase Complex
2.
Sci Bull (Beijing) ; 67(3): 299-314, 2022 02 15.
Article in English | MEDLINE | ID: mdl-36546079

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of pathologies, ranging from steatosis to nonalcoholic steatohepatitis (NASH). The factors promoting the progression of steatosis to NASH are still unclear. Recent studies suggest that mitochondrial lipid composition is critical in NASH development. Here, we showed that CDP-DAG synthase 2 (Cds2) was downregulated in genetic or diet-induced NAFLD mouse models. Liver-specific deficiency of Cds2 provoked hepatic steatosis, inflammation and fibrosis in five-week-old mice. CDS2 is enriched in mitochondria-associated membranes (MAMs), and hepatic Cds2 deficiency impaired mitochondrial function and decreased mitochondrial PE levels. Overexpression of phosphatidylserine decarboxylase (PISD) alleviated the NASH-like phenotype in Cds2f/f;AlbCre mice and abnormal mitochondrial morphology and function caused by CDS2 deficiency in hepatocytes. Additionally, dietary supplementation with an agonist of peroxisome proliferator-activated receptor alpha (PPARα) attenuated mitochondrial defects and ameliorated the NASH-like phenotype in Cds2f/f;AlbCre mice. Finally, Cds2 overexpression protected against high-fat diet-induced hepatic steatosis and obesity. Thus, Cds2 modulates mitochondrial function and NASH development.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Mice , Diacylglycerol Cholinephosphotransferase , Diet, High-Fat , Fibrosis , Mitochondria/pathology , Non-alcoholic Fatty Liver Disease/genetics
3.
Nat Commun ; 12(1): 6877, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824276

ABSTRACT

AGPATs (1-acylglycerol-3-phosphate O-acyltransferases) catalyze the acylation of lysophosphatidic acid to form phosphatidic acid (PA), a key step in the glycerol-3-phosphate pathway for the synthesis of phospholipids and triacylglycerols. AGPAT2 is the only AGPAT isoform whose loss-of-function mutations cause a severe form of human congenital generalized lipodystrophy. Paradoxically, AGPAT2 deficiency is known to dramatically increase the level of its product, PA. Here, we find that AGPAT2 deficiency impairs the biogenesis and growth of lipid droplets. We show that AGPAT2 deficiency compromises the stability of CDP-diacylglycerol (DAG) synthases (CDSs) and decreases CDS activity in both cell lines and mouse liver. Moreover, AGPAT2 and CDS1/2 can directly interact and form functional complexes, which promote the metabolism of PA along the CDP-DAG pathway of phospholipid synthesis. Our results provide key insights into the regulation of metabolic flux during lipid synthesis and suggest substrate channelling at a major branch point of the glycerol-3-phosphate pathway.


Subject(s)
Acyltransferases/metabolism , Cytidine Diphosphate Diglycerides/metabolism , Diacylglycerol Cholinephosphotransferase/metabolism , Fatty Acids/metabolism , Acyltransferases/deficiency , Animals , Biosynthetic Pathways , Cell Line , Diacylglycerol Cholinephosphotransferase/deficiency , Humans , Lipid Droplets/metabolism , Lipogenesis , Liver/metabolism , Mice , Multienzyme Complexes , Oleic Acid/metabolism , Phosphatidic Acids/metabolism
4.
Front Cell Dev Biol ; 8: 615856, 2020.
Article in English | MEDLINE | ID: mdl-33425923

ABSTRACT

Membrane contact sites (MCSs), regions where the membranes of two organelles are closely apposed, play critical roles in inter-organelle communication, such as lipid trafficking, intracellular signaling, and organelle biogenesis and division. First identified as "fraction X" in the early 90s, MCSs are now widely recognized to facilitate local lipid synthesis and inter-organelle lipid transfer, which are important for maintaining cellular lipid homeostasis. In this review, we discuss lipid metabolism and related cellular and physiological functions in MCSs. We start with the characteristics of lipid synthesis and breakdown at MCSs. Then we focus on proteins involved in lipid synthesis and turnover at these sites. Lastly, we summarize the cellular function of lipid metabolism at MCSs beyond mere lipid homeostasis, including the physiological meaning and relevance of MCSs regarding systemic lipid metabolism. This article is part of an article collection entitled: Coupling and Uncoupling: Dynamic Control of Membrane Contacts.

5.
Cell Res ; 28(4): 448-461, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29540758

ABSTRACT

Programmed cell death (PCD) is a fundamental biological process. Deficiency in MOSAIC DEATH 1 (MOD1), a plastid-localized enoyl-ACP reductase, leads to the accumulation of reactive oxygen species (ROS) and PCD, which can be suppressed by mitochondrial complex I mutations, indicating a signal from chloroplasts to mitochondria. However, this signal remains to be elucidated. In this study, through cloning and analyzing a series of mod1 suppressors, we reveal a comprehensive organelle communication pathway that regulates the generation of mitochondrial ROS and triggers PCD. We show that mutations in PLASTIDIAL NAD-DEPENDENT MALATE DEHYDROGENASE (plNAD-MDH), chloroplastic DICARBOXYLATE TRANSPORTER 1 (DiT1) and MITOCHONDRIAL MALATE DEHYDROGENASE 1 (mMDH1) can each rescue the ROS accumulation and PCD phenotypes in mod1, demonstrating a direct communication from chloroplasts to mitochondria via the malate shuttle. Further studies demonstrate that these elements play critical roles in the redox homeostasis and plant growth under different photoperiod conditions. Moreover, we reveal that the ROS level and PCD are significantly increased in malate-treated HeLa cells, which can be dramatically attenuated by knockdown of the human gene MDH2, an ortholog of Arabidopsis mMDH1. These results uncover a conserved malate-induced PCD pathway in plant and animal systems, revolutionizing our understanding of the communication between organelles.


Subject(s)
Arabidopsis/cytology , Arabidopsis/metabolism , Chloroplasts/metabolism , Malates/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Apoptosis , Arabidopsis Proteins/metabolism , Biological Transport , Cell Death , Malate Dehydrogenase/metabolism
6.
Mol Metab ; 9: 131-140, 2018 03.
Article in English | MEDLINE | ID: mdl-29361497

ABSTRACT

OBJECTIVES: Activation of the bile acid (BA) receptors farnesoid X receptor (FXR) or G protein-coupled bile acid receptor (GPBAR1; TGR5) improves metabolic homeostasis. In this study, we aim to determine the impact of pharmacological activation of bile acid receptors by INT-767 on reversal of diet-induced metabolic disorders, and the relative contribution of FXR vs. TGR5 to INT-767's effects on metabolic parameters. METHODS: Wild-type (WT), Tgr5-/-, Fxr-/-, Apoe-/- and Shp-/- mice were used to investigate whether and how BA receptor activation by INT-767, a semisynthetic agonist for both FXR and TGR5, could reverse diet-induced metabolic disorders. RESULTS: INT-767 reversed HFD-induced obesity dependent on activation of both TGR5 and FXR and also reversed the development of atherosclerosis and non-alcoholic fatty liver disease (NAFLD). Mechanistically, INT-767 improved hypercholesterolemia by activation of FXR and induced thermogenic genes via activation of TGR5 and/or FXR. Furthermore, INT-767 inhibited several lipogenic genes and de novo lipogenesis in the liver via activation of FXR. We identified peroxisome proliferation-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (CEBPα) as novel FXR-regulated genes. FXR inhibited PPARγ expression by inducing small heterodimer partner (SHP) whereas the inhibition of CEBPα by FXR was SHP-independent. CONCLUSIONS: BA receptor activation can reverse obesity, NAFLD, and atherosclerosis by specific activation of FXR or TGR5. Our data suggest that, compared to activation of FXR or TGR5 only, dual activation of both FXR and TGR5 is a more attractive strategy for treatment of common metabolic disorders.


Subject(s)
Bile Acids and Salts/therapeutic use , Hypercholesterolemia/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, G-Protein-Coupled/agonists , Animals , Bile Acids and Salts/pharmacology , Diet, High-Fat/adverse effects , Hep G2 Cells , Humans , Hypercholesterolemia/etiology , Hypercholesterolemia/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/etiology , Obesity/metabolism
7.
Sci Rep ; 7(1): 17845, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259301

ABSTRACT

Atherosclerotic cardiovascular disease is a leading cause of death in the western world. Increased plasma triglyceride and cholesterol levels are major risk factors for this disease. Carboxylesterase 1 (Ces1/Ces1g) has been shown to play a role in metabolic control. So far, the role of mouse Ces1/Ces1g deficiency in atherosclerosis is not elucidated. We generated Ces1/Ces1g -/- mice. Compared to wild-type mice, Ces1/Ces1g -/- mice had reduced plasma cholesterol levels. We then generated Ces1g -/- Ldlr -/- double knockout (DKO) mice, which were fed a Western diet for 16 weeks. Compared to Ldlr -/- mice, DKO mice displayed decreased plasma cholesterol and TG levels and reduced atherosclerotic lesions. Interestingly, knockdown of hepatic Ces1/Ces1g in Apoe -/- mice resulted in hyperlipidemia and exacerbated Western diet-induced atherogenesis. Mechanistically, global inactivation of Ces1/Ces1g inhibited intestinal cholesterol and fat absorption and Niemann-Pick C1 like 1 expression, and increased macrophage cholesterol efflux by inducing ATP-binding cassette subfamily A member 1 (ABCA1) and ABCG1. Ces1/Ces1g ablation also promoted M2 macrophage polarization and induced hepatic cholesterol 7α-hydroxylase and sterol 12α-hydroxylase expression. In conclusion, global loss of Ces1/Ces1g protects against the development of atherosclerosis by inhibiting intestinal cholesterol and triglyceride absorption and promoting macrophage cholesterol efflux.


Subject(s)
Atherosclerosis/metabolism , Carboxylic Ester Hydrolases/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Atherosclerosis/blood , Cholesterol/blood , Cholesterol/metabolism , Disease Models, Animal , Hyperlipidemias/blood , Hyperlipidemias/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Triglycerides/blood
8.
Hepatology ; 64(4): 1072-85, 2016 10.
Article in English | MEDLINE | ID: mdl-27359351

ABSTRACT

UNLABELLED: Activation of farnesoid X receptor (FXR) markedly attenuates development of atherosclerosis in animal models. However, the underlying mechanism is not well elucidated. Here, we show that the FXR agonist, obeticholic acid (OCA), increases fecal cholesterol excretion and macrophage reverse cholesterol transport (RCT) dependent on activation of hepatic FXR. OCA does not increase biliary cholesterol secretion, but inhibits intestinal cholesterol absorption. OCA markedly inhibits hepatic cholesterol 7α-hydroxylase (Cyp7a1) and sterol 12α-hydroxylase (Cyp8b1) partly through inducing small heterodimer partner, leading to reduced bile acid pool size and altered bile acid composition, with the α/ß-muricholic acid proportion in bile increased by 2.6-fold and taurocholic acid (TCA) level reduced by 71%. Overexpression of Cyp8b1 or concurrent overexpression of Cyp7a1 and Cyp8b1 normalizes TCA level, bile acid composition, and intestinal cholesterol absorption. CONCLUSION: Activation of FXR inhibits intestinal cholesterol absorption by modulation of bile acid pool size and composition, thus leading to increased RCT. Targeting hepatic FXR and/or bile acids may be useful for boosting RCT and preventing the development of atherosclerosis. (Hepatology 2016;64:1072-1085).


Subject(s)
Bile Acids and Salts/chemistry , Cholesterol/metabolism , Intestinal Absorption , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Biological Transport , Mice , Mice, Inbred C57BL
9.
Sci Rep ; 6: 24277, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27075303

ABSTRACT

The liver is a major organ that controls hepatic and systemic homeostasis. Dysregulation of liver metabolism may cause liver injury. Previous studies have demonstrated that carboxylesterase 1 (CES1) regulates hepatic triglyceride metabolism and protects against liver steatosis. In the present study, we investigated whether CES1 played a role in the development of alcoholic liver disease (ALD) and methionine and choline-deficient (MCD) diet-induced liver injury. Both hepatocyte nuclear factor 4α (HNF4α) and CES1 were markedly reduced in patients with alcoholic steatohepatitis. Alcohol repressed both HNF4α and CES1 expression in primary hepatocytes. HNF4α regulated CES1 expression by directly binding to the proximal promoter of CES1. Global inactivation of CES1 aggravated alcohol- or MCD diet-induced liver inflammation and liver injury, likely as a result of increased production of acetaldehyde and reactive oxygen species and mitochondrial dysfunctions. Knockdown of hepatic CES1 exacerbated ethanol-induced steatohepatitis. These data indicate that CES1 plays a crucial role in protection against alcohol- or MCD diet-induced liver injury.


Subject(s)
Alcohols/toxicity , Carboxylic Ester Hydrolases/metabolism , Chemical and Drug Induced Liver Injury/pathology , Diet/adverse effects , Fatty Liver, Alcoholic/pathology , Hepatocyte Nuclear Factor 4/metabolism , Animals , Gene Expression Regulation , Hepatocytes/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Binding
10.
Nat Commun ; 6: 7466, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26100857

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases, but its underlying mechanism is poorly understood. Here we show that hepatocyte nuclear factor 4α (HNF4α), a liver-enriched nuclear hormone receptor, is markedly inhibited, whereas miR-34a is highly induced in patients with non-alcoholic steatohepatitis, diabetic mice and mice fed a high-fat diet. miR-34a is essential for HNF4α expression and regulates triglyceride accumulation in human and murine hepatocytes. miR-34a inhibits very low-density lipoprotein secretion and promotes liver steatosis and hypolipidemia in an HNF4α-dependent manner. As a result, increased miR-34a or reduced HNF4α expression in the liver attenuates the development of atherosclerosis in Apoe(-/-) or Ldlr(-/-) mice. These data indicate that the miR-34a-HNF4α pathway is activated under common conditions of metabolic stress and may have a role in the pathogenesis of NAFLD and in regulating plasma lipoprotein metabolism. Targeting this pathway may represent a novel approach for the treatment of NAFLD.


Subject(s)
Hepatocyte Nuclear Factor 4/genetics , Lipid Metabolism/genetics , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/genetics , Stress, Physiological/genetics , Triglycerides/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Hep G2 Cells , Hepatocyte Nuclear Factor 4/metabolism , Humans , Lipoproteins/metabolism , Liver/metabolism , Mice, Knockout , MicroRNAs/metabolism , Middle Aged , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, LDL/genetics
11.
PLoS One ; 9(10): e109663, 2014.
Article in English | MEDLINE | ID: mdl-25285996

ABSTRACT

Metabolic syndrome, characterized by obesity, hyperglycemia, dyslipidemia and hypertension, increases the risks for cardiovascular disease, diabetes and stroke. Carboxylesterase 1 (CES1) is an enzyme that hydrolyzes triglycerides and cholesterol esters, and is important for lipid metabolism. Our previous data show that over-expression of mouse hepatic CES1 lowers plasma glucose levels and improves insulin sensitivity in diabetic ob/ob mice. In the present study, we determined the physiological role of hepatic CES1 in glucose homeostasis. Hepatic CES1 expression was reduced by fasting but increased in diabetic mice. Treatment of mice with glucose induced hepatic CES1 expression. Consistent with the in vivo study, glucose stimulated CES1 promoter activity and increased acetylation of histone 3 and histone 4 in the CES1 chromatin. Knockdown of ATP-citrate lyase (ACL), an enzyme that regulates histone acetylation, abolished glucose-mediated histone acetylation in the CES1 chromatin and glucose-induced hepatic CES1 expression. Finally, knockdown of hepatic CES1 significantly increased postprandial blood glucose levels. In conclusion, the present study uncovers a novel glucose-CES1-glucose pathway which may play an important role in regulating postprandial blood glucose levels.


Subject(s)
Blood Glucose/metabolism , Carboxylic Ester Hydrolases/metabolism , Glucose/pharmacology , Liver/enzymology , Postprandial Period , ATP Citrate (pro-S)-Lyase/metabolism , Acetylation/drug effects , Animals , Chromatin/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Histones/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Nutritional Status
12.
Hepatology ; 59(5): 1761-1771, 2014 May.
Article in English | MEDLINE | ID: mdl-24038130

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is one of the major health concerns worldwide. Farnesoid X receptor (FXR) is considered a therapeutic target for treatment of NAFLD. However, the mechanism by which activation of FXR lowers hepatic triglyceride (TG) levels remains unknown. Here we investigated the role of hepatic carboxylesterase 1 (CES1) in regulating both normal and FXR-controlled lipid homeostasis. Overexpression of hepatic CES1 lowered hepatic TG and plasma glucose levels in both wild-type and diabetic mice. In contrast, knockdown of hepatic CES1 increased hepatic TG and plasma cholesterol levels. These effects likely resulted from the TG hydrolase activity of CES1, with subsequent changes in fatty acid oxidation and/or de novo lipogenesis. Activation of FXR induced hepatic CES1, and reduced the levels of hepatic and plasma TG as well as plasma cholesterol in a CES1-dependent manner. CONCLUSION: Hepatic CES1 plays a critical role in regulating both lipid and carbohydrate metabolism and FXR-controlled lipid homeostasis.


Subject(s)
Carboxylic Ester Hydrolases/physiology , Homeostasis , Lipid Metabolism , Liver/enzymology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Cholesterol/blood , Fatty Acids/metabolism , Lipogenesis , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Protein 1/physiology , Triglycerides/metabolism
13.
Mol Endocrinol ; 26(2): 272-80, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22261820

ABSTRACT

Farnesoid X receptor (FXR) is known to play important regulatory roles in bile acid, lipid, and carbohydrate metabolism. Aged (>12 months old) Fxr(-/-) mice also develop spontaneous liver carcinomas. In this report, we used three mouse models to investigate the role of FXR deficiency in obesity. As compared with low-density lipoprotein receptor (Ldlr) knockout (Ldlr(-/-)) mice, the Ldlr(-/-)Fxr(-/-) double-knockout mice were highly resistant to diet-induced obesity, which was associated with increased expression of genes involved in energy metabolism in the skeletal muscle and brown adipose tissue. Such a striking effect of FXR deficiency on obesity on an Ldlr(-/-) background led us to investigate whether FXR deficiency alone is sufficient to affect obesity. As compared with wild-type mice, Fxr(-/-) mice showed resistance to diet-induced weight gain. Interestingly, only female Fxr(-/-) mice showed significant resistance to diet-induced obesity, which was accompanied by increased energy expenditure in these mice. Finally, we determined the effect of FXR deficiency on obesity in a genetically obese and diabetic mouse model. We generated ob(-/-)Fxr(-/-) mice that were deficient in both Leptin and Fxr. On a chow diet, ob(-/-)Fxr(-/-) mice gained less body weight and had reduced body fat mass as compared with ob/ob mice. In addition, we observed liver carcinomas in 43% of young (<11 months old) Ob(-/-)Fxr(-/-) mice. Together these data indicate that loss of FXR prevents diet-induced or genetic obesity and accelerates liver carcinogenesis under diabetic conditions.


Subject(s)
Carcinoma/genetics , Diet, High-Fat/adverse effects , Liver Neoplasms/genetics , Obesity/etiology , Receptors, Cytoplasmic and Nuclear/deficiency , Adipose Tissue, Brown/pathology , Adiposity/genetics , Animals , Carcinoma/etiology , Cell Transformation, Neoplastic/genetics , Dietary Fats/metabolism , Energy Metabolism/genetics , Female , Gene Knockout Techniques , Glucose Intolerance/complications , Glucose Intolerance/genetics , Intestinal Absorption , Leptin/deficiency , Leptin/genetics , Liver/pathology , Liver Neoplasms/etiology , Male , Mice , Mice, Knockout , Mice, Obese , Muscle, Skeletal/metabolism , Obesity/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Sex Factors , Weight Gain/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...