Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.455
Filter
1.
Cell Rep ; 43(5): 114238, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38748875

ABSTRACT

Triacylglyceride (TAG) synthesis in the small intestine determines the absorption of dietary fat, but the underlying mechanisms remain to be further studied. Here, we report that the RNA-binding protein HuR (ELAVL1) promotes TAG synthesis in the small intestine. HuR associates with the 3' UTR of Dgat2 mRNA and intron 1 of Mgat2 pre-mRNA. Association of HuR with Dgat2 3' UTR stabilizes Dgat2 mRNA, while association of HuR with intron 1 of Mgat2 pre-mRNA promotes the processing of Mgat2 pre-mRNA. Intestinal epithelium-specific HuR knockout reduces the expression of DGAT2 and MGAT2, thereby reducing the dietary fat absorption through TAG synthesis and mitigating high-fat-diet (HFD)-induced non-alcoholic fatty liver disease (NAFLD) and obesity. Our findings highlight a critical role of HuR in promoting dietary fat absorption.


Subject(s)
Diet, High-Fat , ELAV-Like Protein 1 , Intestinal Absorption , Triglycerides , Triglycerides/metabolism , Triglycerides/biosynthesis , Animals , ELAV-Like Protein 1/metabolism , ELAV-Like Protein 1/genetics , Mice , Diet, High-Fat/adverse effects , Humans , Mice, Inbred C57BL , Male , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/genetics , Obesity/metabolism , Obesity/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , Dietary Fats/metabolism , Dietary Fats/pharmacology , Mice, Knockout , 3' Untranslated Regions/genetics , Acyltransferases
2.
Nat Commun ; 15(1): 3547, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670976

ABSTRACT

Typical plant membranes and storage lipids are comprised of five common fatty acids yet over 450 unusual fatty acids accumulate in seed oils of various plant species. Plant oils are important human and animal nutrients, while some unusual fatty acids such as hydroxylated fatty acids (HFA) are used in the chemical industry (lubricants, paints, polymers, cosmetics, etc.). Most unusual fatty acids are extracted from non-agronomic crops leading to high production costs. Attempts to engineer HFA into crops are unsuccessful due to bottlenecks in the overlapping pathways of oil and membrane lipid synthesis where HFA are not compatible. Physaria fendleri naturally overcomes these bottlenecks through a triacylglycerol (TAG) remodeling mechanism where HFA are incorporated into TAG after initial synthesis. TAG remodeling involves a unique TAG lipase and two diacylglycerol acyltransferases (DGAT) that are selective for different stereochemical and acyl-containing species of diacylglycerol within a synthesis, partial degradation, and resynthesis cycle. The TAG lipase interacts with DGAT1, localizes to the endoplasmic reticulum (with the DGATs) and to puncta around the lipid droplet, likely forming a TAG remodeling metabolon near the lipid droplet-ER junction. Each characterized DGAT and TAG lipase can increase HFA accumulation in engineered seed oils.


Subject(s)
Diacylglycerol O-Acyltransferase , Fatty Acids , Plant Oils , Triglycerides , Triglycerides/metabolism , Triglycerides/biosynthesis , Plant Oils/metabolism , Plant Oils/chemistry , Diacylglycerol O-Acyltransferase/metabolism , Diacylglycerol O-Acyltransferase/genetics , Fatty Acids/metabolism , Lipase/metabolism , Seeds/metabolism , Endoplasmic Reticulum/metabolism , Plant Proteins/metabolism , Plant Proteins/genetics , Lipid Droplets/metabolism , Plants, Genetically Modified
3.
Mol Microbiol ; 121(6): 1164-1181, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38676355

ABSTRACT

Latent tuberculosis, caused by dormant Mycobacterium tuberculosis (Mtb), poses a threat to global health through the incubation of undiagnosed infections within the community. Dormant Mtb, which is phenotypically tolerant to antibiotics, accumulates triacylglycerol (TAG) utilizing fatty acids obtained from macrophage lipid droplets. TAG is vital to mycobacteria, serving as a cell envelope component and energy reservoir during latency. TAG synthesis occurs by sequential acylation of glycerol-3-phosphate, wherein the second acylation step is catalyzed by acylglycerol-3-phosphate acyltransferase (AGPAT), resulting in the production of phosphatidic acid (PA), a precursor for the synthesis of TAG and various phospholipids. Here, we have characterized a putative acyltransferase of Mtb encoded by Rv3816c. We found that Rv3816c has all four characteristic motifs of AGPAT, exists as a membrane-bound enzyme, and functions as 1-acylglycerol-3-phosphate acyltransferase. The enzyme could transfer the acyl group to acylglycerol-3-phosphate (LPA) from monounsaturated fatty acyl-coenzyme A of chain length 16 or 18 to produce PA. Complementation of Escherichia coli PlsC mutant in vivo by Rv3816c confirmed that it functions as AGPAT. Its active site mutants, H43A and D48A, were incapable of transferring the acyl group to LPA in vitro and were not able to rescue the growth defect of E. coli PlsC mutant in vivo. Identifying Rv3816c as AGPAT and comparing its properties with other AGPAT homologs is not only a step toward understanding the TAG biosynthesis in mycobacteria but has the potential to explore it as a drug target.


Subject(s)
Mycobacterium tuberculosis , Triglycerides , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Triglycerides/biosynthesis , Triglycerides/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Glycerol-3-Phosphate O-Acyltransferase/genetics , Acyltransferases/metabolism , Acyltransferases/genetics , Acylation , Fatty Acids/metabolism , Fatty Acids/biosynthesis , Phosphatidic Acids/metabolism , Phosphatidic Acids/biosynthesis , Acyl Coenzyme A/metabolism
4.
BMC Plant Biol ; 24(1): 309, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38649801

ABSTRACT

BACKGROUND: Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), belonging to ω-3 long-chain polyunsaturated fatty acids (ω3-LC-PUFAs), are essential components of human diet. They are mainly supplemented by marine fish consumption, although their native producers are oleaginous microalgae. Currently, increasing demand for fish oils is insufficient to meet the entire global needs, which puts pressure on searching for the alternative solutions. One possibility may be metabolic engineering of plants with an introduced enzymatic pathway producing ω3-LC-PUFAs. RESULT: In this study we focused on the acyl-CoA:diacylglycerol acyltransferase2b (PtDGAT2b) from the diatom Phaeodactylum tricornutum, an enzyme responsible for triacylglycerol (TAG) biosynthesis via acyl-CoA-dependent pathway. Gene encoding PtDGAT2b, incorporated into TAG-deficient yeast strain H1246, was used to confirm its activity and conduct biochemical characterization. PtDGAT2b exhibited a broad acyl-CoA preference with both di-16:0-DAG and di-18:1-DAG, whereas di-18:1-DAG was favored. The highest preference for acyl donors was observed for 16:1-, 10:0- and 12:0-CoA. PtDGAT2b also very efficiently utilized CoA-conjugated ω-3 LC-PUFAs (stearidonic acid, eicosatetraenoic acid and EPA). Additionally, verification of the potential role of PtDGAT2b in planta, through its transient expression in tobacco leaves, indicated increased TAG production with its relative amount increasing to 8%. Its co-expression with the gene combinations aimed at EPA biosynthesis led to, beside elevated TAG accumulation, efficient accumulation of EPA which constituted even 25.1% of synthesized non-native fatty acids (9.2% of all fatty acids in TAG pool). CONCLUSIONS: This set of experiments provides a comprehensive biochemical characterization of DGAT enzyme from marine microalgae. Additionally, this study elucidates that PtDGAT2b can be used successfully in metabolic engineering of plants designed to obtain a boosted TAG level, enriched not only in ω-3 LC-PUFAs but also in medium-chain and ω-7 fatty acids.


Subject(s)
Diacylglycerol O-Acyltransferase , Diatoms , Nicotiana , Diatoms/genetics , Diatoms/enzymology , Diatoms/metabolism , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Nicotiana/genetics , Nicotiana/enzymology , Nicotiana/metabolism , Acyl Coenzyme A/metabolism , Plants, Genetically Modified , Triglycerides/biosynthesis , Triglycerides/metabolism , Eicosapentaenoic Acid/biosynthesis , Eicosapentaenoic Acid/metabolism , Fatty Acids, Omega-3/biosynthesis , Fatty Acids, Omega-3/metabolism , Metabolic Engineering
5.
J Biol Chem ; 300(1): 105587, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38141768

ABSTRACT

The Saccharomyces cerevisiae Nem1-Spo7 protein phosphatase complex dephosphorylates and thereby activates Pah1 at the nuclear/endoplasmic reticulum membrane. Pah1, a phosphatidate phosphatase catalyzing the dephosphorylation of phosphatidate to produce diacylglycerol, is one of the most highly regulated enzymes in lipid metabolism. The diacylglycerol produced in the lipid phosphatase reaction is utilized for the synthesis of triacylglycerol that is stored in lipid droplets. Disruptions of the Nem1-Spo7/Pah1 phosphatase cascade cause a plethora of physiological defects. Spo7, the regulatory subunit of the Nem1-Spo7 complex, is required for the Nem1 catalytic function and interacts with the acidic tail of Pah1. Spo7 contains three conserved homology regions (CR1-3) that are important for the interaction with Nem1, but its region for the interaction with Pah1 is unknown. Here, by deletion and site-specific mutational analyses of Spo7, we revealed that the C-terminal basic tail (residues 240-259) containing five arginine and two lysine residues is important for the Nem1-Spo7 complex-mediated dephosphorylation of Pah1 and its cellular function (triacylglycerol synthesis, lipid droplet formation, maintenance of nuclear/endoplasmic reticulum membrane morphology, and cell growth at elevated temperatures). The glutaraldehyde cross-linking analysis of synthetic peptides indicated that the Spo7 basic tail interacts with the Pah1 acidic tail. This work advances our understanding of the Spo7 function and the Nem1-Spo7/Pah1 phosphatase cascade in yeast lipid synthesis.


Subject(s)
Membrane Proteins , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Diglycerides/biosynthesis , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Triglycerides/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism
6.
FEBS Lett ; 598(10): 1226-1234, 2024 May.
Article in English | MEDLINE | ID: mdl-38140812

ABSTRACT

Triglycerides constitute an inert storage form for fatty acids deposited in lipid droplets and are mobilized to provide metabolic energy or membrane building blocks. The biosynthesis of triglycerides is highly conserved within eukaryotes and normally involves the sequential esterification of activated fatty acids with a glycerol backbone. Some eukaryotes, however, can also use cellular membrane lipids as direct fatty acid donors for triglyceride synthesis. The biological significance of a pathway that generates triglycerides at the expense of organelle membranes has remained elusive. Here we review current knowledge on how cells use membrane lipids as fatty acid donors for triglyceride synthesis and discuss the hypothesis that a primary function of this pathway is to regulate membrane lipid remodeling and organelle function.


Subject(s)
Membrane Lipids , Organelles , Triglycerides , Triglycerides/metabolism , Triglycerides/biosynthesis , Humans , Animals , Membrane Lipids/metabolism , Organelles/metabolism , Fatty Acids/metabolism , Fatty Acids/biosynthesis , Cell Membrane/metabolism
7.
Nature ; 621(7977): 171-178, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648867

ABSTRACT

Triacylglycerols (TAGs) are the main source of stored energy in the body, providing an important substrate pool for mitochondrial beta-oxidation. Imbalances in the amount of TAGs are associated with obesity, cardiac disease and various other pathologies1,2. In humans, TAGs are synthesized from excess, coenzyme A-conjugated fatty acids by diacylglycerol O-acyltransferases (DGAT1 and DGAT2)3. In other organisms, this activity is complemented by additional enzymes4, but whether such alternative pathways exist in humans remains unknown. Here we disrupt the DGAT pathway in haploid human cells and use iterative genetics to reveal an unrelated TAG-synthesizing system composed of a protein we called DIESL (also known as TMEM68, an acyltransferase of previously unknown function) and its regulator TMX1. Mechanistically, TMX1 binds to and controls DIESL at the endoplasmic reticulum, and loss of TMX1 leads to the unconstrained formation of DIESL-dependent lipid droplets. DIESL is an autonomous TAG synthase, and expression of human DIESL in Escherichia coli endows this organism with the ability to synthesize TAG. Although both DIESL and the DGATs function as diacylglycerol acyltransferases, they contribute to the cellular TAG pool under specific conditions. Functionally, DIESL synthesizes TAG at the expense of membrane phospholipids and maintains mitochondrial function during periods of extracellular lipid starvation. In mice, DIESL deficiency impedes rapid postnatal growth and affects energy homeostasis during changes in nutrient availability. We have therefore identified an alternative TAG biosynthetic pathway driven by DIESL under potent control by TMX1.


Subject(s)
Acyltransferases , Triglycerides , Animals , Humans , Mice , Acyltransferases/metabolism , Coenzyme A/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Escherichia coli/metabolism , Homeostasis , Triglycerides/biosynthesis , Energy Metabolism , Nutrients/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism
8.
J Lipid Res ; 64(3): 100339, 2023 03.
Article in English | MEDLINE | ID: mdl-36737040

ABSTRACT

Treatment with acetyl-CoA carboxylase inhibitors (ACCi) in nonalcoholic steatohepatitis (NASH) may increase plasma triglycerides (TGs), with variable changes in apoB concentrations. ACC is rate limiting in de novo lipogenesis and regulates fatty acid oxidation, making it an attractive therapeutic target in NASH. Our objectives were to determine the effects of the ACCi, firsocostat, on production rates of plasma LDL-apoB in NASH and the effects of combined therapy with fenofibrate. Metabolic labeling with heavy water and tandem mass spectrometric analysis of LDL-apoB enrichments was performed in 16 NASH patients treated with firsocostat for 12 weeks and in 29 NASH subjects treated with firsocostat and fenofibrate for 12 weeks. In NASH on firsocostat, plasma TG increased significantly by 17% from baseline to week 12 (P = 0.0056). Significant increases were also observed in LDL-apoB fractional replacement rate (baseline to week 12: 31 ± 20.2 to 46 ± 22.6%/day, P = 0.03) and absolute synthesis rate (ASR) (30.4-45.2 mg/dl/day, P = 0.016) but not plasma apoB concentrations. The effect of firsocostat on LDL-apoB ASR was restricted to patients with cirrhosis (21.0 ± 9.6 at baseline and 44.2 ± 17 mg/dl/day at week 12, P = 0.002, N = 8); noncirrhotic patients did not change (39.8 ± 20.8 and 46.3 ± 14.8 mg/dl/day, respectively, P = 0.51, N = 8). Combination treatment with fenofibrate and firsocostat prevented increases in plasma TG, LDL-apoB fractional replacement rate, and ASR. In summary, in NASH with cirrhosis, ACCi treatment increases LDL-apoB100 production rate and this effect can be prevented by concurrent fenofibrate therapy.


Subject(s)
Acetyl-CoA Carboxylase , Fenofibrate , Liver Cirrhosis , Non-alcoholic Fatty Liver Disease , Humans , Acetyl-CoA Carboxylase/antagonists & inhibitors , Apolipoproteins B/biosynthesis , Fenofibrate/therapeutic use , Fenofibrate/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Triglycerides/biosynthesis , Triglycerides/blood , Cholesterol, LDL/biosynthesis
9.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36768334

ABSTRACT

Triacylglycerol (TG) biosynthesis is an important metabolic process for intracellular storage of surplus energy, intestinal dietary fat absorption, attenuation of lipotoxicity, lipid transportation, lactation and signal transduction in mammals. Transmembrane protein 68 (TMEM68) is an endoplasmic reticulum (ER)-anchored acyltransferase family member of unknown function. In the current study we show that overexpression of TMEM68 promotes TG accumulation and lipid droplet (LD) formation in a conserved active sites-dependent manner. Quantitative targeted lipidomic analysis showed that diacylglycerol (DG), free fatty acid (FFA) and TG levels were increased by TMEM68 expression. In addition, TMEM68 overexpression affected the levels of several glycerophospholipids, such as phosphatidylcholine, phosphatidylethanolamine and phosphatidylinositol, as well as sterol ester contents. TMEM68 exhibited monoacylglycerol acyltransferase (MGAT) and diacylglycerol acyltransferase (DGAT) activities dependent on the conserved active sites in an in vitro assay. The expression of lipogenesis genes, including DGATs, fatty acid synthesis-related genes and peroxisome proliferator-activated receptor γ was upregulated in TMEM68-overexpressing cells. These results together demonstrate for the first time that TMEM68 functions as an acyltransferase and affects lipogenic gene expression, glycerolipid metabolism and TG storage in mammalian cells.


Subject(s)
Diacylglycerol O-Acyltransferase , Lipogenesis , Membrane Proteins , Triglycerides , Animals , Female , Humans , Acyltransferases/genetics , Acyltransferases/metabolism , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Lipogenesis/genetics , Lipogenesis/physiology , Mammals/metabolism , Triglycerides/biosynthesis , Triglycerides/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism
10.
Sci Rep ; 12(1): 2390, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149744

ABSTRACT

Studies on 3T3-L1 cells and HepG2 hepatocytes have shown that phosphatidic acid phosphohydrolase1 (LPIN1) plays a key role in adipogenesis, acting as a co-activator of peroxisome proliferator-activated receptor gamma coactivator 1a (PGC-1a) to regulate fatty acid metabolism. However, the functional role and regulatory mechanism of LPIN1 gene in milk fat synthesis of buffalo are still unknown. In this study, overexpression of buffalo LPIN1 gene transfected with recombinant fusion expression vector significantly increased the expression of AGPAT6, DGAT1, DGAT2, GPAM and BTN1A1 genes involved in triglyceride (TAG) synthesis and secretion, as well as PPARG and SREBF1 genes regulating fatty acid metabolism in the buffalo mammary epithelial cells (BMECs), while the lentivirus-mediated knockdown of buffalo LPIN1 dramatically decreased the relative mRNA abundance of these genes. Correspondingly, total cellular TAG content in the BMECs increased significantly after LPIN1 overexpression, but decreased significantly after LPIN1 knockdown. In addition, the overexpression or knockdown of PPARG also enhanced or reduced the expression of LPIN1 and the transcriptional activity of its promoter. The core region of buffalo LPIN1 promoter spans from - 666 bp to + 42 bp, and two PPAR response elements (PPREs: PPRE1 and PPRE2) were identified in this region. Site mutagenesis analysis showed that PPARG directly regulated the transcription of buffalo LPIN1 by binding to the PPRE1 and PPRE2 on its core promoter. The results here reveal that the LPIN1 gene is involved in the milk fat synthesis of BMECs, and one of the important pathways is to participate in this process through direct transcriptional regulation of PPARG, which in turn significantly affects the content of TAG in BMECs.


Subject(s)
Buffaloes/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/metabolism , PPAR gamma/metabolism , Phosphatidate Phosphatase/genetics , Triglycerides/biosynthesis , Animals , Buffaloes/genetics , Female , Gene Expression Regulation , Milk/metabolism , PPAR gamma/genetics , Phosphatidate Phosphatase/metabolism , Transcription, Genetic
11.
Cornea ; 41(1): 95-105, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34483274

ABSTRACT

PURPOSE: PGF2α analogs are commonly used to treat glaucoma and are associated with higher rates of meibomian gland dysfunction (MGD). The purpose of this study was to evaluate the physiological effects of PGF2α and PGE2 on immortalized human meibomian gland epithelial cells (HMGECs). METHODS: HMGECs were immunostained for the 4 PGE2 receptors (EP1, EP2, EP3, and EP4) and 1 PGF2α receptor (FP) and imaged. Rosiglitazone-differentiated HMGECs were exposed to PGF2α and PGE2 (10-9 to 10-6 M) for 3 hours. Cell viability was assessed by an adenosine triphosphate-based luminescent assay, and lipid extracts were analyzed for cholesteryl esters (CEs), wax esters (WEs), and triacylglycerols (TAGs) by ESI-MSMSALL in positive ion mode by a Triple TOF 5600 Mass Spectrometer using SCIEX LipidView 1.3. RESULTS: HMGECs expressed 3 PGE2 receptors (EP1, EP2, and EP4) and the 1 PGF2α receptor (FP). Neither PGE2 nor PGF2α showed signs of cytotoxicity at any of the concentrations tested. WEs were not detected from any of the samples, but both CEs and TAGs exhibited a diverse and dynamic profile. PGE2 suppressed select CEs (CE 22:1, CE 26:0, CE 28:1, and CE 30:1). PGF2α dose dependently increased several CEs (CE 20:2, CE 20:1, CE 22:1, and CE 24:0) yet decreased others. Both prostaglandins led to nonspecific TAG remodeling. CONCLUSIONS: PGE2 and PGF2α showed minimal effect on HMGEC viability. PGF2α influences lipid expression greater than PGE2 and may do so by interfering with meibocyte differentiation. This work may provide insight into the mechanism of MGD development in patients with glaucoma treated with PGF2α analogs.


Subject(s)
Cholesterol Esters/biosynthesis , Epithelial Cells/metabolism , Meibomian Glands/cytology , Receptors, Prostaglandin E, EP2 Subtype/biosynthesis , Receptors, Prostaglandin/biosynthesis , Triglycerides/biosynthesis , Cell Count , Cells, Cultured , Epithelial Cells/cytology , Humans , Immunohistochemistry , Mass Spectrometry , Meibomian Glands/metabolism
12.
PLoS One ; 16(12): e0256973, 2021.
Article in English | MEDLINE | ID: mdl-34879063

ABSTRACT

We recently identified a nuclear-encoded miRNA (miR-181c) in cardiomyocytes that can translocate into mitochondria to regulate mitochondrial gene mt-COX1 and influence obesity-induced cardiac dysfunction through the mitochondrial pathway. Because liver plays a pivotal role during obesity, we hypothesized that miR-181c might contribute to the pathophysiological complications associated with obesity. Therefore, we used miR-181c/d-/- mice to study the role of miR-181c in hepatocyte lipogenesis during diet-induced obesity. The mice were fed a high-fat (HF) diet for 26 weeks, during which indirect calorimetric measurements were made. Quantitative PCR (qPCR) was used to examine the expression of genes involved in lipid synthesis. We found that miR-181c/d-/- mice were not protected against all metabolic consequences of HF exposure. After 26 weeks, the miR-181c/d-/- mice had a significantly higher body fat percentage than did wild-type (WT) mice. Glucose tolerance tests showed hyperinsulinemia and hyperglycemia, indicative of insulin insensitivity in the miR-181c/d-/- mice. miR-181c/d-/- mice fed the HF diet had higher serum and liver triglyceride levels than did WT mice fed the same diet. qPCR data showed that several genes regulated by isocitrate dehydrogenase 1 (IDH1) were more upregulated in miR-181c/d-/- liver than in WT liver. Furthermore, miR-181c delivered in vivo via adeno-associated virus attenuated the lipogenesis by downregulating these same lipid synthesis genes in the liver. In hepatocytes, miR-181c regulates lipid biosynthesis by targeting IDH1. Taken together, the data indicate that overexpression of miR-181c can be beneficial for various lipid metabolism disorders.


Subject(s)
Diet, High-Fat/adverse effects , Hepatocytes/metabolism , Lipogenesis , Liver/metabolism , MicroRNAs/metabolism , Obesity , Triglycerides , Animals , Lipogenesis/drug effects , Lipogenesis/genetics , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Obesity/chemically induced , Obesity/genetics , Obesity/metabolism , Triglycerides/biosynthesis , Triglycerides/genetics
13.
Cells ; 10(11)2021 11 17.
Article in English | MEDLINE | ID: mdl-34831427

ABSTRACT

MicroRNAs (miRNAs), a class of small, non-coding RNA molecules, play an important role in the posttranscriptional regulation of gene expression, thereby influencing important cellular functions. In adipocytes, miRNAs show import regulatory features and are described to influence differentiation as well as metabolic, endocrine, and inflammatory functions. We previously identified miR-27a being upregulated under inflammatory conditions in human adipocytes and aimed to elucidate its function in adipocyte biology. Both strands of miR-27a, miR-27a-3p and -5p, were downregulated during the adipogenic differentiation of Simpson-Golabi-Behmel syndrome (SGBS) cells, human multipotent adipose-derived stem cells (hMADS), and human primary adipose-derived stromal cells (hASCs). Using miRNA-mimic transfection, we observed that miR-27a-3p is a crucial regulator of adipogenesis, while miR-27a-5p did not alter the differentiation capacity in SGBS cells. In silico screening predicted lipoprotein lipase (LPL) and peroxisome proliferator activated receptor γ (PPARγ) as potential targets of miR-27a-3p. The downregulation of both genes was verified in vitro, and the interaction of miR-27-3p with target sites in the 3' UTRs of both genes was confirmed via a miRNA-reporter-gene assay. Here, the knockdown of LPL did not interfere with adipogenic differentiation, while PPARγ knockdown decreased adipogenesis significantly, suggesting that miR-27-3p exerts its inhibitory effect on adipogenesis by repressing PPARγ. Taken together, we identified and validated a crucial role for miR-27a-3p in human adipogenesis played by targeting the essential adipogenic transcription factor PPARγ. Though we confirmed LPL as an additional target of miR-27a-3p, it does not appear to be involved in regulating human adipogenesis. Thereby, our findings call the conclusions drawn from previous studies, which identified LPL as a crucial regulator for murine and human adipogenesis, into question.


Subject(s)
Adipogenesis/genetics , MicroRNAs/metabolism , Base Sequence , Biomarkers/metabolism , Female , Gene Expression Regulation , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , MicroRNAs/genetics , Middle Aged , PPAR gamma/metabolism , Triglycerides/biosynthesis
14.
J Lipid Res ; 62: 100152, 2021.
Article in English | MEDLINE | ID: mdl-34808194

ABSTRACT

Testosterone is a hormone essential for male reproductive function. It is produced primarily by Leydig cells in the testicle through activation of steroidogenic acute regulatory protein and a series of steroidogenic enzymes, including a cytochrome P450 side-chain cleavage enzyme (cytochome P450 family 11 subfamily A member 1), 17α-hydroxylase (cytochrome P450 family 17 subfamily A member 1), and 3ß-hydroxysteroid dehydrogenase. These steroidogenic enzymes are mainly regulated at the transcriptional level, and their expression is increased by the nuclear receptor 4A1. However, the effect on Leydig cell function of a small molecule-activating ligand, amodiaquine (AQ), is unknown. We found that AQ effectively and significantly increased testosterone production in TM3 and primary Leydig cells through enhanced expression of steroidogenic acute regulatory protein, cytochome P450 family 11 subfamily A member 1, cytochrome P450 family 17 subfamily A member 1, and 3ß-hydroxysteroid dehydrogenase. Concurrently, AQ dose-dependently increased the expression of 3-hydroxy-3-methylglutaryl-CoA reductase, a key enzyme in the cholesterol synthesis pathway, through induction of the transcriptional and DNA-binding activities of nuclear receptor 4A1, contributing to increased cholesterol synthesis in Leydig cells. Furthermore, AQ increased the expression of fatty acid synthase and diacylglycerol acyltransferase and potentiated de novo synthesis of fatty acids and triglycerides (TGs). Lipidomics profiling further confirmed a significant elevation of intracellular lipid and TG levels by AQ in Leydig cells. These results demonstrated that AQ effectively promotes testosterone production and de novo synthesis of cholesterol and TG in Leydig cells, indicating that AQ may be beneficial for treating patients with Leydig cell dysfunction and subsequent testosterone deficiency.


Subject(s)
Amodiaquine/pharmacology , Cholesterol/biosynthesis , Leydig Cells/drug effects , Testosterone/biosynthesis , Triglycerides/biosynthesis , Animals , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL
15.
Plant J ; 108(6): 1735-1753, 2021 12.
Article in English | MEDLINE | ID: mdl-34643970

ABSTRACT

Light quantity and quality affect many aspects of plant growth and development. However, few reports have addressed the molecular connections between seed oil accumulation and light conditions, especially dense shade. Shade-avoiding plants can redirect plant resources into extension growth at the expense of leaf and root expansion in an attempt to reach areas containing richer light. Here, we report that tung tree seed oil accumulation is suppressed by dense shade during the rapid oil accumulation phase. Transcriptome analysis confirmed that oil accumulation suppression due to dense shade was attributed to reduced expression of fatty acid and triacylglycerol biosynthesis-related genes. Through weighted gene co-expression network analysis, we identified 32 core transcription factors (TFs) specifically upregulated in densely shaded seeds during the rapid oil accumulation period. Among these, VfHB21, a class I homeodomain leucine zipper TF, was shown to suppress expression of FAD2 and FADX, two key genes related to α-eleostearic acid, by directly binding to HD-ZIP I/II motifs in their respective promoter regions. VfHB21 also binds to similar motifs in the promoters of VfWRI1 and VfDGAT2, two additional key seed lipid regulatory/biosynthetic genes. Functional conservation of HB21 during plant evolution was demonstrated by the fact that AtWRI1, AtSAD1, and AtFAD2 were downregulated in VfHB21-overexpressor lines of transgenic Arabidopsis, with concomitant seed oil reduction, and the fact that AtHB21 expression also was induced by shade. This study reveals some of the regulatory mechanisms that specifically control tung tree seed oil biosynthesis and more broadly regulate plant storage carbon partitioning in response to dense shade conditions.


Subject(s)
Euphorbiaceae/metabolism , Plant Proteins/genetics , Seeds/metabolism , Triglycerides/biosynthesis , Arabidopsis/genetics , Arabidopsis/metabolism , Euphorbiaceae/genetics , Fatty Acid Desaturases/genetics , Gene Expression Regulation, Plant , Leucine Zippers , Light , Linolenic Acids/genetics , Linolenic Acids/metabolism , Phylogeny , Plant Growth Regulators/genetics , Plant Growth Regulators/metabolism , Plant Oils/metabolism , Plant Proteins/metabolism , Plants, Genetically Modified , Promoter Regions, Genetic , Seeds/genetics , Seeds/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , Trees , Triglycerides/genetics
16.
Front Immunol ; 12: 722735, 2021.
Article in English | MEDLINE | ID: mdl-34603294

ABSTRACT

Lipid metabolism plays a complex and dynamic role in host-pathogen interaction during Mycobacterium tuberculosis infection. While bacterial lipid metabolism is key to the success of the pathogen, the host also offers a lipid rich environment in the form of necrotic caseous granulomas, making this association beneficial for the pathogen. Accumulation of the neutral lipid triglyceride, as lipid droplets within the cellular cuff of necrotic granulomas, is a peculiar feature of pulmonary tuberculosis. The role of triglyceride synthesis in the TB granuloma and its impact on the disease outcome has not been studied in detail. Here, we identified diacylglycerol O-acyltransferase 1 (DGAT1) to be essential for accumulation of triglyceride in necrotic TB granulomas using the C3HeB/FeJ murine model of infection. Treatment of infected mice with a pharmacological inhibitor of DGAT1 (T863) led to reduction in granuloma triglyceride levels and bacterial burden. A decrease in bacterial burden was associated with reduced neutrophil infiltration and degranulation, and a reduction in several pro-inflammatory cytokines including IL1ß, TNFα, IL6, and IFNß. Triglyceride lowering impacted eicosanoid production through both metabolic re-routing and via transcriptional control. Our data suggests that manipulation of lipid droplet homeostasis may offer a means for host directed therapy in Tuberculosis.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Granuloma/drug therapy , Mycobacterium tuberculosis/immunology , Triglycerides/biosynthesis , Tuberculosis/drug therapy , Animals , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Granuloma/microbiology , Granuloma/pathology , Host-Pathogen Interactions , Male , Mice , Mice, Inbred C3H , Mycobacterium tuberculosis/drug effects , Neutrophil Infiltration , Tuberculosis/complications , Tuberculosis/immunology , Tuberculosis/microbiology
17.
Nat Commun ; 12(1): 5323, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493722

ABSTRACT

The role of intestine clock in energy homeostasis remains elusive. Here we show that mice with Bmal1 specifically deleted in the intestine (Bmal1iKO mice) have a normal phenotype on a chow diet. However, on a high-fat diet (HFD), Bmal1iKO mice are protected against development of obesity and related abnormalities such as hyperlipidemia and fatty livers. These metabolic phenotypes are attributed to impaired lipid resynthesis in the intestine and reduced fat secretion. Consistently, wild-type mice fed a HFD during nighttime (with a lower BMAL1 expression) show alleviated obesity compared to mice fed ad libitum. Mechanistic studies uncover that BMAL1 transactivates the Dgat2 gene (encoding the triacylglycerol synthesis enzyme DGAT2) via direct binding to an E-box in the promoter, thereby promoting dietary fat absorption. Supporting these findings, intestinal deficiency of Rev-erbα, a known BMAL1 repressor, enhances dietary fat absorption and exacerbates HFD-induced obesity and comorbidities. Moreover, small-molecule targeting of REV-ERBα/BMAL1 by SR9009 ameliorates HFD-induced obesity in mice. Altogether, intestine clock functions as an accelerator in dietary fat absorption and targeting intestinal BMAL1 may be a promising approach for management of metabolic diseases induced by excess fat intake.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Rhythm/genetics , Diacylglycerol O-Acyltransferase/genetics , Fatty Liver/genetics , Hyperlipidemias/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Obesity/genetics , ARNTL Transcription Factors/deficiency , Animals , Diacylglycerol O-Acyltransferase/metabolism , Diet, High-Fat/adverse effects , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Gene Expression Regulation , Homeostasis/drug effects , Homeostasis/genetics , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/prevention & control , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/prevention & control , Promoter Regions, Genetic , Protein Binding , Pyrrolidines/pharmacology , Signal Transduction , Thiophenes/pharmacology , Triglycerides/biosynthesis
18.
Food Funct ; 12(18): 8681-8693, 2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34351342

ABSTRACT

This study examined the impact of chitobiose (GlcN)2 and chitotriose (GlcN)3 on lipid accumulation modification and their inhibitory functionalities. (GlcN)2 and (GlcN)3 significantly inhibited the total cholesterol (TC), triglyceride (TG), and low-density lipid cholesterol (LDL-c) levels in the liver of the ob/ob-/- mice fed a non-high-fat diet. This phenomenon was associated with a reduction in the mRNA and protein expression of TG synthesis and fatty acid uptake-related signaling, significantly affecting the cluster of differentiation 36 (CD36) and diacylglycerol acyltransferase 2 (DGAT2). Furthermore, the CD36 and DGAT2 genes were overexpressed by constructing a plasmid and transfecting it into HepG2 cells, after which the phenotypic traits of lipid accumulation were assessed in vitro. Consequently, it was evident that (GlcN)2 and (GlcN)3 reduced the overexpression of these proteins and relieved cellular lipid accumulation. In conclusion, these results indicated that (GlcN)2 and (GlcN)3 acted positively against NAFLD while regulating steatosis in the non-high-fat diet NAFLD model. The potential NAFLD treatment strategies, such as targeting CD36 and DGAT2 signaling, could provide scientific insight into further applying food-derived ingredients to reduce the risk of high-fat metabolism.


Subject(s)
CD36 Antigens/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Disaccharides/administration & dosage , Fatty Acids/administration & dosage , Non-alcoholic Fatty Liver Disease/therapy , Triglycerides/biosynthesis , Trisaccharides/administration & dosage , Animals , CD36 Antigens/genetics , Diacylglycerol O-Acyltransferase/genetics , Diet , Dietary Fats/administration & dosage , Gene Expression Regulation , Hep G2 Cells , Humans , Lipid Metabolism/genetics , Lipids/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction , Simvastatin/pharmacology
19.
Life Sci ; 283: 119866, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34352257

ABSTRACT

AIMS: Morphine, a commonly used drug for anesthesia, affects lipid metabolism in different tissues, but the mechanism is currently unclear. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme responsible for the first step of triglyceride (TG) hydrolysis. Here we aim to investigate whether ATGL phosphorylation is involved in morphine-induced TG accumulation. MAIN METHODS: Oil red O staining and TG content analysis were used to detect the effect of morphine on lipid storage. A series of ATGL phosphoamino acid site mutant plasmids were constructed by gene synthesis and transfected to HL-1 cells to evaluate the phosphorylation levels of ATGL phosphoamino acid in morphine-treated HL-1 cells with immunoprecipitation and immunoblotting assay. KEY FINDINGS: Morphine acute treatment induced excessive accumulation of TG and decreased the phosphorylation level of ATGL Ser406 in HL-1 cells. Of note, the phosphorylation positive mutation of ATGL Ser406 to aspartic acid effectively reversed morphine-induced excessive accumulation of TG in HL-1 cells. SIGNIFICANCE: This discovery will help to fully understand the lipid regulation function of morphine in a new scope. In addition, it will expand the phosphorylation research of ATGL more comprehensively and provide powerful clues for lipid metabolism regulation.


Subject(s)
Lipase/metabolism , Morphine/pharmacology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Triglycerides/biosynthesis , Animals , Cell Line , Male , Mice , Morphine/pharmacokinetics , Myocardium/pathology , Myocytes, Cardiac/pathology , Phosphorylation/drug effects
20.
Int J Mol Sci ; 22(12)2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34208774

ABSTRACT

Bile acids (BA) play a significant role in the pathophysiology of nonalcoholic steatohepatitis (NASH). The present study evaluates the modulation of bile acid metabolomics by atorvastatin, a cholesterol-lowering agent commonly used to treat cardiovascular complications accompanying NASH. NASH was induced in mice by 24 weeks of consuming a high-saturated fat, high-fructose, and high-cholesterol diet (F), with atorvastatin administered orally (20 mg/kg/day) during the last three weeks. Biochemical and histological analyses confirmed the effectiveness of the F diet in inducing NASH. Untreated NASH animals had significantly reduced biliary secretion of BA and increased fecal excretion of BA via decreased apical sodium-dependent bile salt transporter (Asbt)-mediated reabsorption. Atorvastatin decreased liver steatosis and inflammation in NASH animals consistently with a reduction in crucial lipogenic enzyme stearoyl-coenzyme A (CoA) desaturase-1 and nuclear factor kappa light chain enhancer of activated B-cell pro-inflammatory signaling, respectively. In this group, atorvastatin also uniformly enhanced plasma concentration, biliary secretion and fecal excretion of the secondary BA, deoxycholic acid (DCA). However, in the chow diet-fed animals, atorvastatin decreased plasma concentrations of BA, and reduced BA biliary secretions. These changes stemmed primarily from the increased fecal excretion of BA resulting from the reduced Asbt-mediated BA reabsorption in the ileum and suppression of synthesis in the liver. In conclusion, our results reveal that atorvastatin significantly modulates BA metabolomics by altering their intestinal processing and liver synthesis in control and NASH mice.


Subject(s)
Atorvastatin/pharmacology , Bile Acids and Salts/metabolism , Homeostasis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Biomarkers , Diet, High-Fat , Disease Models, Animal , Liver/metabolism , Mice , Models, Biological , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Triglycerides/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...