Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Bull Math Biol ; 85(9): 82, 2023 08 06.
Article in English | MEDLINE | ID: mdl-37544001

ABSTRACT

Fatty acids (FAs) are crucial energy metabolites, signalling molecules, and membrane building blocks for a wide range of organisms. Adipose triglyceride lipase (ATGL) is the first and presumingly most crucial regulator of FA release from triacylglycerols (TGs) stored within cytosolic lipid droplets. However, besides the function of releasing FAs by hydrolysing TGs into diacylglycerols (DGs), ATGL also promotes the transacylation reaction of two DG molecules into one TG and one monoacylglycerol molecule. To date, it is unknown whether DG transacylation is a coincidental byproduct of ATGL-mediated lipolysis or whether it is physiologically relevant. Experimental evidence is scarce since both, hydrolysis and transacylation, rely on the same active site of ATGL and always occur in parallel in an ensemble of molecules. This paper illustrates the potential roles of transacylation. It shows that, depending on the kinetic parameters but also on the state of the hydrolytic machinery, transacylation can increase or decrease downstream products up to 80% respectively 30%. We provide an extensive asymptotic analysis including quasi-steady-state approximations (QSSA) with higher order correction terms and provide numerical simulation. We also argue that when assessing the validity of QSSAs one should include parameter sensitivity derivatives. Our results suggest that the transacylation function of ATGL is of biological relevance by providing feedback options and altogether stability to the lipolytic machinery in adipocytes.


Subject(s)
Lipase , Lipolysis , Lipolysis/physiology , Lipase/metabolism , Mathematical Concepts , Models, Biological , Adipocytes , Fatty Acids/metabolism , Triglycerides/metabolism
2.
Mol Metab ; 71: 101705, 2023 05.
Article in English | MEDLINE | ID: mdl-36907508

ABSTRACT

OBJECTIVE: In brown adipose tissue (iBAT), the balance between lipid/glucose uptake and lipolysis is tightly regulated by insulin signaling. Downstream of the insulin receptor, PDK1 and mTORC2 phosphorylate AKT, which activates glucose uptake and lysosomal mTORC1 signaling. The latter requires the late endosomal/lysosomal adaptor and MAPK and mTOR activator (LAMTOR/Ragulator) complex, which serves to translate the nutrient status of the cell to the respective kinase. However, the role of LAMTOR in metabolically active iBAT has been elusive. METHODS: Using an AdipoqCRE-transgenic mouse line, we deleted LAMTOR2 (and thereby the entire LAMTOR complex) in adipose tissue (LT2 AKO). To examine the metabolic consequences, we performed metabolic and biochemical studies in iBAT isolated from mice housed at different temperatures (30 °C, room temperature and 5 °C), after insulin treatment, or in fasted and refed condition. For mechanistic studies, mouse embryonic fibroblasts (MEFs) lacking LAMTOR 2 were analyzed. RESULTS: Deletion of the LAMTOR complex in mouse adipocytes resulted in insulin-independent AKT hyperphosphorylation in iBAT, causing increased glucose and fatty acid uptake, which led to massively enlarged lipid droplets. As LAMTOR2 was essential for the upregulation of de novo lipogenesis, LAMTOR2 deficiency triggered exogenous glucose storage as glycogen in iBAT. These effects are cell autonomous, since AKT hyperphosphorylation was abrogated by PI3K inhibition or by deletion of the mTORC2 component Rictor in LAMTOR2-deficient MEFs. CONCLUSIONS: We identified a homeostatic circuit for the maintenance of iBAT metabolism that links the LAMTOR-mTORC1 pathway to PI3K-mTORC2-AKT signaling downstream of the insulin receptor.


Subject(s)
Proto-Oncogene Proteins c-akt , Receptor, Insulin , Mice , Animals , Receptor, Insulin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adipose Tissue, Brown/metabolism , Fibroblasts/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Insulin/metabolism , Mice, Transgenic , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Nutrients , Homeostasis , Glucose/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proteins/metabolism
3.
Front Genet ; 13: 913030, 2022.
Article in English | MEDLINE | ID: mdl-35734423

ABSTRACT

Active thermogenic adipocytes avidly consume energy substrates like fatty acids and glucose to maintain body temperature upon cold exposure. Despite strong evidence for the involvement of brown adipose tissue (BAT) in controlling systemic energy homeostasis upon nutrient excess, it is unclear how the activity of brown adipocytes is regulated in times of nutrient scarcity. Therefore, this study aimed to scrutinize factors that modulate BAT activity to balance thermogenic and energetic needs upon simultaneous fasting and cold stress. For an unbiased view, we performed transcriptomic and miRNA sequencing analyses of BAT from acutely fasted (24 h) mice under mild cold exposure. Combining these data with in-depth bioinformatic analyses and in vitro gain-of-function experiments, we define a previously undescribed axis of p53 inducing miR-92a-1-5p transcription that is highly upregulated by fasting in thermogenic adipocytes. p53, a fasting-responsive transcription factor, was previously shown to control genes involved in the thermogenic program and miR-92a-1-5p was found to negatively correlate with human BAT activity. Here, we identify fructose transporter Slc2a5 as one direct downstream target of this axis and show that fructose can be taken up by and metabolized in brown adipocytes. In sum, this study delineates a fasting-induced pathway involving p53 that transactivates miR-92a-1-5p, which in turn decreases Slc2a5 expression, and suggests fructose as an energy substrate in thermogenic adipocytes.

4.
J Biol Chem ; 297(4): 101206, 2021 10.
Article in English | MEDLINE | ID: mdl-34543623

ABSTRACT

Adipose triglyceride lipase (ATGL) plays a key role in intracellular lipolysis, the mobilization of stored triacylglycerol. This work provides an important basis for generating reproducible and detailed data on the hydrolytic and transacylation activities of ATGL. We generated full-length and C-terminally truncated ATGL variants fused with various affinity tags and analyzed their expression in different hosts, namely E.coli, the insect cell line Sf9, and the mammalian cell line human embryonic kidney 293T. Based on this screen, we expressed a fusion protein of ATGL covering residues M1-D288 flanked with N-terminal and C-terminal purification tags. Using these fusions, we identified key steps in expression and purification protocols, including production in the E. coli strain ArcticExpress (DE3) and removal of copurified chaperones. The resulting purified ATGL variant demonstrated improved lipolytic activity compared with previously published data, and it could be stimulated by the coactivator protein comparative gene identification-58 and inhibited by the protein G0/G1 switch protein 2. Shock freezing and storage did not affect the basal activity but reduced coactivation of ATGL by comparative gene identification 58. In vitro, the truncated ATGL variant demonstrated acyl-CoA-independent transacylation activity when diacylglycerol was offered as substrate, resulting in the formation of fatty acid as well as triacylglycerol and monoacylglycerol. However, the ATGL variant showed neither hydrolytic activity nor transacylation activity upon offering of monoacylglycerol as substrate. To understand the role of ATGL in different physiological contexts, it is critical for future studies to identify all its different functions and to determine under what conditions these activities occur.


Subject(s)
Gene Expression , Lipase , Acylation , Animals , HEK293 Cells , Humans , Hydrolysis , Lipase/biosynthesis , Lipase/chemistry , Lipase/genetics , Lipase/isolation & purification , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sf9 Cells , Spodoptera
5.
Cell ; 184(13): 3502-3518.e33, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34048700

ABSTRACT

Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of ß-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3.


Subject(s)
Adipose Tissue, Brown/metabolism , Constitutive Androstane Receptor/metabolism , Lipolysis , Receptors, G-Protein-Coupled/metabolism , Thermogenesis , Adipocytes/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cold Temperature , Dietary Fats/pharmacology , Humans , Mice, Inbred C57BL , Phenotype , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Sympathetic Nervous System/metabolism , Transcription, Genetic
6.
Commun Biol ; 4(1): 323, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33692445

ABSTRACT

Modulation of adipocyte lipolysis represents an attractive approach to treat metabolic diseases. Lipolysis mainly depends on two enzymes: adipose triglyceride lipase and hormone-sensitive lipase (HSL). Here, we investigated the short- and long-term impact of adipocyte HSL on energy homeostasis using adipocyte-specific HSL knockout (AHKO) mice. AHKO mice fed high-fat-diet (HFD) progressively developed lipodystrophy accompanied by excessive hepatic lipid accumulation. The increased hepatic triglyceride deposition was due to induced de novo lipogenesis driven by increased fatty acid release from adipose tissue during refeeding related to defective insulin signaling in adipose tissue. Remarkably, the fatty liver of HFD-fed AHKO mice reversed with advanced age. The reversal of fatty liver coincided with a pronounced lipodystrophic phenotype leading to blunted lipolytic activity in adipose tissue. Overall, we demonstrate that impaired adipocyte HSL-mediated lipolysis affects systemic energy homeostasis in AHKO mice, whereby with older age, these mice reverse their fatty liver despite advanced lipodystrophy.


Subject(s)
Adipocytes/enzymology , Energy Metabolism , Fatty Liver/enzymology , Lipodystrophy/enzymology , Lipolysis , Liver/metabolism , Sterol Esterase/deficiency , Adipocytes/pathology , Age Factors , Animals , Blood Glucose/metabolism , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/pathology , Insulin/metabolism , Lipodystrophy/genetics , Lipodystrophy/pathology , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , Sterol Esterase/genetics , Time Factors
7.
Sci Transl Med ; 13(580)2021 02 10.
Article in English | MEDLINE | ID: mdl-33568522

ABSTRACT

Heart failure with preserved ejection fraction (HFpEF) is a highly prevalent and intractable form of cardiac decompensation commonly associated with diastolic dysfunction. Here, we show that diastolic dysfunction in patients with HFpEF is associated with a cardiac deficit in nicotinamide adenine dinucleotide (NAD+). Elevating NAD+ by oral supplementation of its precursor, nicotinamide, improved diastolic dysfunction induced by aging (in 2-year-old C57BL/6J mice), hypertension (in Dahl salt-sensitive rats), or cardiometabolic syndrome (in ZSF1 obese rats). This effect was mediated partly through alleviated systemic comorbidities and enhanced myocardial bioenergetics. Simultaneously, nicotinamide directly improved cardiomyocyte passive stiffness and calcium-dependent active relaxation through increased deacetylation of titin and the sarcoplasmic reticulum calcium adenosine triphosphatase 2a, respectively. In a long-term human cohort study, high dietary intake of naturally occurring NAD+ precursors was associated with lower blood pressure and reduced risk of cardiac mortality. Collectively, these results suggest NAD+ precursors, and especially nicotinamide, as potential therapeutic agents to treat diastolic dysfunction and HFpEF in humans.


Subject(s)
Heart Failure , Animals , Cohort Studies , Heart Failure/drug therapy , Humans , Mice , Mice, Inbred C57BL , Niacinamide/pharmacology , Niacinamide/therapeutic use , Rats , Rats, Inbred Dahl , Stroke Volume
8.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33372146

ABSTRACT

Branched esters of palmitic acid and hydroxy stearic acid are antiinflammatory and antidiabetic lipokines that belong to a family of fatty acid (FA) esters of hydroxy fatty acids (HFAs) called FAHFAs. FAHFAs themselves belong to oligomeric FA esters, known as estolides. Glycerol-bound FAHFAs in triacylglycerols (TAGs), named TAG estolides, serve as metabolite reservoir of FAHFAs mobilized by lipases upon demand. Here, we characterized the involvement of two major metabolic lipases, adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL), in TAG estolide and FAHFA degradation. We synthesized a library of 20 TAG estolide isomers with FAHFAs varying in branching position, chain length, saturation grade, and position on the glycerol backbone and developed an in silico mass spectra library of all predicted catabolic intermediates. We found that ATGL alone or coactivated by comparative gene identification-58 efficiently liberated FAHFAs from TAG estolides with a preference for more compact substrates where the estolide branching point is located near the glycerol ester bond. ATGL was further involved in transesterification and remodeling reactions leading to the formation of TAG estolides with alternative acyl compositions. HSL represented a much more potent estolide bond hydrolase for both TAG estolides and free FAHFAs. FAHFA and TAG estolide accumulation in white adipose tissue of mice lacking HSL argued for a functional role of HSL in estolide catabolism in vivo. Our data show that ATGL and HSL participate in the metabolism of estolides and TAG estolides in distinct manners and are likely to affect the lipokine function of FAHFAs.


Subject(s)
Lipase/metabolism , Sterol Esterase/metabolism , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Esters/chemistry , Fatty Acids/metabolism , Female , HEK293 Cells , Humans , Lipolysis/physiology , Metabolism/physiology , Mice , Mice, Knockout , Palmitic Acid/metabolism , Stearic Acids/metabolism , Triglycerides/metabolism
9.
Mol Cell Endocrinol ; 512: 110869, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32439414

ABSTRACT

Cold exposure activates brown and brown-like adipocytes that dissipate large amounts of glucose and fatty acids via uncoupling protein 1 (UCP1) to drive non-shivering thermogenesis (NST). Evidence for the existence of these thermogenic adipocytes in adult humans gave rise to a renaissance in research on brown adipose tissue, establishing it as linchpin of energy homeostasis and metabolic health. Besides low ambient temperature, shortage or excess of food affect thermoregulation. Upon high caloric meals thermogenic adipocytes burn excess calories and maintain energy balance. In contrast, in conditions of nutrient deprivation, counter-regulatory mechanisms prevent thermogenic adipocytes from "wasting" energy substrates that need to be conserved. In this review, we discuss cell-autonomous mechanisms, metabolites, and hormones that modify NST in response to nutrient fluctuations. In particular, we focus on how thermogenic adipocytes balance thermogenesis with systemic energy homeostasis during fasting periods.


Subject(s)
Adipocytes/physiology , Cold Temperature , Fasting/metabolism , Thermogenesis/physiology , Adaptation, Physiological/physiology , Adipose Tissue, Brown/physiology , Animals , Body Temperature Regulation/physiology , Energy Metabolism/physiology , Humans , Hypothermia/metabolism , Hypothermia/physiopathology
10.
J Lipid Res ; 61(7): 995-1003, 2020 07.
Article in English | MEDLINE | ID: mdl-32350080

ABSTRACT

Bis(monoacylglycero)phosphate (BMP), also known as lysobisphosphatidic acid, is a phospholipid that promotes lipid sorting in late endosomes/lysosomes by activating lipid hydrolases and lipid transfer proteins. Changes in the cellular BMP content therefore reflect an altered metabolic activity of the endolysosomal system. Surprisingly, little is known about the physiological regulation of BMP. In this study, we investigated the effects of nutritional and metabolic factors on BMP profiles of whole tissues and parenchymal and nonparenchymal cells. Tissue samples were obtained from fed, fasted, 2 h refed, and insulin-treated mice, as well as from mice housed at 5°C, 22°C, or 30°C. These tissues exhibited distinct BMP profiles that were regulated by the nutritional state in a tissue-specific manner. Insulin treatment was not sufficient to mimic refeeding-induced changes in tissue BMP levels, indicating that BMP metabolism is regulated by other hormonal or nutritional factors. Tissue fractionation experiments revealed that fasting drastically elevates BMP levels in hepatocytes and pancreatic cells. Furthermore, we observed that the BMP content in brown adipose tissue strongly depends on housing temperatures. In conclusion, our observations suggest that BMP concentrations adapt to the metabolic state in a tissue- and cell-type-specific manner in mice. Drastic changes observed in hepatocytes, pancreatic cells, and brown adipocytes suggest that BMP plays a role in the functional adaption to nutrient starvation and ambient temperature.


Subject(s)
Lysophospholipids/metabolism , Lysosomes/metabolism , Monoglycerides/metabolism , Animals , Endosomes/metabolism , Macrophages/cytology , Mice
11.
Metabolites ; 10(4)2020 Apr 10.
Article in English | MEDLINE | ID: mdl-32290093

ABSTRACT

The catabolism of intracellular triacylglycerols (TAGs) involves the activity of cytoplasmic and lysosomal enzymes. Cytoplasmic TAG hydrolysis, commonly termed lipolysis, is catalyzed by the sequential action of three major hydrolases, namely adipose triglyceride lipase, hormone-sensitive lipase, and monoacylglycerol lipase. All three enzymes interact with numerous protein binding partners that modulate their activity, cellular localization, or stability. Deficiencies of these auxiliary proteins can lead to derangements in neutral lipid metabolism and energy homeostasis. In this review, we summarize the composition and the dynamics of the complex lipolytic machinery we like to call "lipolysome".

12.
Cardiovasc Res ; 116(2): 339-352, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31166588

ABSTRACT

AIMS: Lipotoxic cardiomyopathy in diabetic and obese patients typically encompasses increased cardiac fatty acid (FA) uptake eventually surpassing the mitochondrial oxidative capacity. Lowering FA utilization via inhibition of lipolysis represents a strategy to counteract the development of lipotoxic heart dysfunction. However, defective cardiac triacylglycerol (TAG) catabolism and FA oxidation in humans (and mice) carrying mutated ATGL alleles provokes lipotoxic heart dysfunction questioning a therapeutic approach to decrease cardiac lipolysis. Interestingly, decreased lipolysis via cardiac overexpression of Perilipin 5 (Plin5), a binding partner of ATGL, is compatible with normal heart function and lifespan despite massive cardiac lipid accumulation. Herein, we decipher mechanisms that protect Plin5 transgenic mice from the development of heart dysfunction. METHODS AND RESULTS: We generated mice with cardiac-specific overexpression of Plin5 encoding a serine-155 to alanine exchange (Plin5-S155A) of the protein kinase A phosphorylation site, which has been suggested as a prerequisite to stimulate lipolysis and may play a crucial role in the preservation of heart function. Plin5-S155A mice showed a substantial increase in cardiac TAG and ceramide levels, which was comparable to mice overexpressing non-mutated Plin5. Lipid accumulation was compatible with normal heart function even under mild stress. Plin5-S155A mice showed reduced cardiac FA oxidation but normal ATP production and changes in the Plin5-S155A phosphoproteome compared to Plin5 transgenic mice. Interestingly, mitochondrial recruitment of dynamin-related protein 1 (Drp1) was markedly reduced in cardiac muscle of Plin5-S155A and Plin5 transgenic mice accompanied by decreased phosphorylation of mitochondrial fission factor, a mitochondrial receptor of Drp1. CONCLUSIONS: This study suggests that low cardiac lipolysis is associated with reduced mitochondrial fission and may represent a strategy to combat the development of lipotoxic heart dysfunction.


Subject(s)
Adipose Tissue/metabolism , Heart Diseases/prevention & control , Intracellular Signaling Peptides and Proteins/metabolism , Lipolysis , Mitochondria, Heart/metabolism , Mitochondrial Dynamics , Muscle Proteins/metabolism , Myocytes, Cardiac/metabolism , Adenosine Triphosphate/metabolism , Adipose Tissue/pathology , Animals , COS Cells , Ceramides/metabolism , Chlorocebus aethiops , Disease Models, Animal , Dynamins/metabolism , Fatty Acids/metabolism , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/metabolism , Mice, Mutant Strains , Mitochondria, Heart/pathology , Mitochondrial Proteins/metabolism , Muscle Proteins/genetics , Mutation , Myocytes, Cardiac/pathology , Oxidation-Reduction , Phosphorylation , Rats , Signal Transduction , Triglycerides/metabolism
13.
Diabetes ; 69(3): 300-312, 2020 03.
Article in English | MEDLINE | ID: mdl-31806624

ABSTRACT

Branched esters of palmitic acid and hydroxystearic acid (PAHSA) are anti-inflammatory and antidiabetic lipokines that connect glucose and lipid metabolism. We aimed to characterize involvement of the 5-PAHSA regioisomer in the adaptive metabolic response of white adipose tissue (WAT) to cold exposure (CE) in mice, exploring the cross talk between glucose utilization and lipid metabolism. CE promoted local production of 5- and 9-PAHSAs in WAT. Metabolic labeling of de novo lipogenesis (DNL) using 2H2O revealed that 5-PAHSA potentiated the effects of CE and stimulated triacylglycerol (TAG)/fatty acid (FA) cycling in WAT through impacting lipogenesis and lipolysis. Adipocyte lipolytic products were altered by 5-PAHSA through selective FA re-esterification. The impaired lipolysis in global adipose triglyceride lipase (ATGL) knockout mice reduced free PAHSA levels and uncovered a metabolite reservoir of TAG-bound PAHSAs (TAG estolides) in WAT. Utilization of 13C isotope tracers and dynamic metabolomics documented that 5-PAHSA primes adipocytes for glucose metabolism in a different way from insulin, promoting DNL and impeding TAG synthesis. In summary, our data reveal new cellular and physiological mechanisms underlying the beneficial effects of 5-PAHSA and its relation to insulin action in adipocytes and independently confirm a PAHSA metabolite reservoir linked to ATGL-mediated lipolysis.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, White/metabolism , Glucose/metabolism , Lipase/genetics , Palmitic Acid/metabolism , Stearic Acids/metabolism , Triglycerides/metabolism , Animals , Carbon Isotopes , Cold Temperature , Deuterium Oxide , Fatty Acids/metabolism , Lipase/metabolism , Lipogenesis/genetics , Lipolysis , Metabolomics , Mice , Mice, Knockout
14.
FASEB J ; 33(12): 13808-13824, 2019 12.
Article in English | MEDLINE | ID: mdl-31638418

ABSTRACT

N-acetylaspartate (NAA) is synthesized by aspartate N-acetyltransferase (gene: Nat8l) from acetyl-coenzyme A and aspartate. In the brain, NAA is considered an important energy metabolite for lipid synthesis. However, the role of NAA in peripheral tissues remained elusive. Therefore, we characterized the metabolic phenotype of knockout (ko) and adipose tissue-specific (ako) Nat8l-ko mice as well as NAA-supplemented mice on various diets. We identified an important role of NAA availability in the brain during adolescence, as 75% of Nat8l-ko mice died on fat-free diet (FFD) after weaning but could be rescued by NAA supplementation. In adult life, NAA deficiency promotes a beneficial metabolic phenotype, as Nat8l-ko and Nat8l-ako mice showed reduced body weight, increased energy expenditure, and improved glucose tolerance on chow, high-fat, and FFDs. Furthermore, Nat8l-deficient adipocytes exhibited increased mitochondrial respiration, ATP synthesis, and an induction of browning. Conversely, NAA-treated wild-type mice showed reduced adipocyte respiration and lipolysis and increased de novo lipogenesis, culminating in reduced energy expenditure, glucose tolerance, and insulin sensitivity. Mechanistically, our data point to a possible role of NAA as modulator of pancreatic insulin secretion and suggest NAA as a critical energy metabolite for adipocyte and whole-body energy homeostasis.-Hofer, D. C., Zirkovits, G., Pelzmann, H. J., Huber, K., Pessentheiner, A. R., Xia, W., Uno, K., Miyazaki, T., Kon, K., Tsuneki, H., Pendl, T., Al Zoughbi, W., Madreiter-Sokolowski, C. T., Trausinger, G., Abdellatif, M., Schoiswohl, G., Schreiber, R., Eisenberg, T., Magnes, C., Sedej, S., Eckhardt, M., Sasahara, M., Sasaoka, T., Nitta, A., Hoefler, G., Graier, W. F., Kratky, D., Auwerx, J., Bogner-Strauss, J. G. N-acetylaspartate availability is essential for juvenile survival on fat-free diet and determines metabolic health.


Subject(s)
Aspartic Acid/analogs & derivatives , Acetyl Coenzyme A/metabolism , Acetyltransferases/metabolism , Adipocytes/metabolism , Animals , Aspartic Acid/metabolism , Brain/metabolism , Diet, Fat-Restricted , Energy Metabolism/physiology , Insulin Resistance/physiology , Lipolysis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism
15.
J Biol Chem ; 294(23): 9118-9133, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31023823

ABSTRACT

Lysosomal acid lipase (LAL) hydrolyzes cholesteryl ester (CE) and retinyl ester (RE) and triglyceride (TG). Mice globally lacking LAL accumulate CE most prominently in the liver. The severity of the CE accumulation phenotype progresses with age and is accompanied by hepatomegaly and hepatic cholesterol crystal deposition. In contrast, hepatic TG accumulation is much less pronounced in these mice, and hepatic RE levels are even decreased. To dissect the functional role of LAL for neutral lipid ester mobilization in the liver, we generated mice specifically lacking LAL in hepatocytes (hep-LAL-ko). On a standard chow diet, hep-LAL-ko mice exhibited increased hepatic CE accumulation but unaltered TG and RE levels. Feeding the hep-LAL-ko mice a vitamin A excess/high-fat diet (VitA/HFD) further increased hepatic cholesterol levels, but hepatic TG and RE levels in these mice were lower than in control mice. Performing in vitro activity assays with lysosome-enriched fractions from livers of mice globally lacking LAL, we detected residual acid hydrolytic activities against TG and RE. Interestingly, this non-LAL acid TG hydrolytic activity was elevated in lysosome-enriched fractions from livers of hep-LAL-ko mice upon VitA/HFD feeding. In conclusion, the neutral lipid ester phenotype in livers from hep-LAL-ko mice indicates that LAL is limiting for CE turnover, but not for TG and RE turnovers. Furthermore, in vitro hydrolase activity assays revealed the existence of non-LAL acid hydrolytic activities for TG and RE. The corresponding acid lipase(s) catalyzing these reactions remains to be identified.


Subject(s)
Cholesterol Esters/metabolism , Diterpenes/metabolism , Liver/metabolism , Sterol Esterase/genetics , Triglycerides/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Cholesterol/blood , Cholesterol/metabolism , Diet, High-Fat , Diterpenes/chemistry , Hepatocytes/cytology , Hepatocytes/metabolism , Lipids/analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Phospholipids/analysis , Sterol Esterase/deficiency , Sterol Esterase/metabolism , Vitamin A/administration & dosage
16.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(6): 880-899, 2019 06.
Article in English | MEDLINE | ID: mdl-30367950

ABSTRACT

Adipose triglyceride lipase (ATGL) has been discovered 14 years ago and revised our view on intracellular triglyceride (TG) mobilization - a process termed lipolysis. ATGL initiates the hydrolysis of TGs to release fatty acids (FAs) that are crucial energy substrates, precursors for the synthesis of membrane lipids, and ligands of nuclear receptors. Thus, ATGL is a key enzyme in whole-body energy homeostasis. In this review, we give an update on how ATGL is regulated on the transcriptional and post-transcriptional level and how this affects the enzymes' activity in the context of neutral lipid catabolism. In depth, we highlight and discuss the numerous physiological functions of ATGL in lipid and energy metabolism. Over more than a decade, different genetic mouse models lacking or overexpressing ATGL in a cell- or tissue-specific manner have been generated and characterized. Moreover, pharmacological studies became available due to the development of a specific murine ATGL inhibitor (Atglistatin®). The identification of patients with mutations in the human gene encoding ATGL and their disease spectrum has underpinned the importance of ATGL in humans. Together, mouse models and human data have advanced our understanding of the physiological role of ATGL in lipid and energy metabolism in adipose and non-adipose tissues, and of the pathophysiological consequences of ATGL dysfunction in mice and men.


Subject(s)
Adipose Tissue/metabolism , Lipase/metabolism , Animals , Energy Metabolism/physiology , Humans , Lipid Metabolism/physiology , Lipids/physiology , Mice
17.
Cell Metab ; 28(4): 644-655.e4, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30033199

ABSTRACT

The coordination of the organ-specific responses regulating systemic energy distribution to replenish lipid stores in acutely activated brown adipose tissue (BAT) remains elusive. Here, we show that short-term cold exposure or acute ß3-adrenergic receptor (ß3AR) stimulation results in secretion of the anabolic hormone insulin. This process is diminished in adipocyte-specific Atgl-/- mice, indicating that lipolysis in white adipose tissue (WAT) promotes insulin secretion. Inhibition of pancreatic ß cells abolished uptake of lipids delivered by triglyceride-rich lipoproteins into activated BAT. Both increased lipid uptake into BAT and whole-body energy expenditure in response to ß3AR stimulation were blunted in mice treated with the insulin receptor antagonist S961 or lacking the insulin receptor in brown adipocytes. In conclusion, we introduce the concept that acute cold and ß3AR stimulation trigger a systemic response involving WAT, ß cells, and BAT, which is essential for insulin-dependent fuel uptake and adaptive thermogenesis.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Cold Temperature , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Lipolysis/physiology , Receptors, Adrenergic, beta-3/metabolism , Adipocytes, Brown/metabolism , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Diet, High-Fat , Dioxoles/pharmacology , Energy Metabolism/physiology , Lipase/metabolism , Lipoproteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides/pharmacology , Receptor, Insulin/antagonists & inhibitors , Thermogenesis/physiology , Triglycerides/metabolism
18.
Cell Rep ; 23(7): 1948-1961, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768196

ABSTRACT

Elevated circulating fatty acids (FAs) contribute to obesity-associated metabolic complications, but the mechanisms by which insulin suppresses lipolysis are poorly understood. We show that α/ß-hydrolase domain-containing 15 (ABHD15) is required for the anti-lipolytic action of insulin in white adipose tissue (WAT). Neither insulin nor glucose treatments can suppress FA mobilization in global and conditional Abhd15-knockout (KO) mice. Accordingly, insulin signaling is impaired in Abhd15-KO adipocytes, as indicated by reduced AKT phosphorylation, glucose uptake, and de novo lipogenesis. In vitro data reveal that ABHD15 associates with and stabilizes phosphodiesterase 3B (PDE3B). Accordingly, PDE3B expression is decreased in the WAT of Abhd15-KO mice, mechanistically explaining increased protein kinase A (PKA) activity, hormone-sensitive lipase (HSL) phosphorylation, and undiminished FA release upon insulin signaling. Ultimately, Abhd15-KO mice develop insulin resistance. Notably, ABHD15 expression is decreased in humans with obesity and diabetes compared to humans with obesity and normal glucose tolerance, identifying ABHD15 as a potential therapeutic target to mitigate insulin resistance.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Insulin Resistance , Insulin/pharmacology , Lipolysis , Membrane Proteins/metabolism , 3T3-L1 Cells , Adipose Tissue, White/metabolism , Animals , Carboxylic Ester Hydrolases/genetics , Diet, High-Fat , Enzyme Stability/drug effects , Fatty Acids/metabolism , Female , Gene Expression Regulation/drug effects , Glucose/metabolism , Humans , Lipolysis/drug effects , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/pathology , Phenotype
19.
Cell Metab ; 26(5): 753-763.e7, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-28988821

ABSTRACT

Fatty acids (FAs) activate and fuel UCP1-mediated non-shivering thermogenesis (NST) in brown adipose tissue (BAT). Release of FAs from intracellular fat stores by adipose triglyceride lipase (ATGL) is considered a key step in NST. Accordingly, the severe cold intolerance of global ATGL knockout (AKO) mice has been attributed to defective BAT lipolysis. Here we show that this conclusion is incorrect. We demonstrate that although the BAT-specific loss of ATGL impairs BAT lipolysis and alters BAT morphology, it does not compromise the ß3-adrenergic thermogenic response or cold-induced NST. Instead, NST depends on nutrient supply or lipolysis in white adipose tissue during fasting, suggesting that circulating energy substrates are sufficient to fuel NST. Cold intolerance in AKO mice is not caused by BAT dysfunction as previously suspected but by severe cardiomyopathy. We conclude that functional NST requires adequate substrate supply and cardiac function, but does not depend on ATGL-mediated lipolysis in BAT.


Subject(s)
Adipose Tissue, Brown/metabolism , Cold Temperature , Lipase/physiology , Lipolysis/physiology , Myocardium/metabolism , Thermogenesis/physiology , Adipose Tissue, White/metabolism , Animals , Eating , Electrocardiography , Fasting , Lipase/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
20.
Cell Metab ; 26(3): 509-522.e6, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877455

ABSTRACT

Cold-induced thermogenesis is an energy-demanding process that protects endotherms against a reduction in ambient temperature. Using non-targeted liquid chromatography-mass spectrometry-based lipidomics, we identified elevated levels of plasma acylcarnitines in response to the cold. We found that the liver undergoes a metabolic switch to provide fuel for brown fat thermogenesis by producing acylcarnitines. Cold stimulates white adipocytes to release free fatty acids that activate the nuclear receptor HNF4α, which is required for acylcarnitine production in the liver and adaptive thermogenesis. Once in circulation, acylcarnitines are transported to brown adipose tissue, while uptake into white adipose tissue and liver is blocked. Finally, a bolus of L-carnitine or palmitoylcarnitine rescues the cold sensitivity seen with aging. Our data highlight an elegant mechanism whereby white adipose tissue provides long-chain fatty acids for hepatic carnitilation to generate plasma acylcarnitines as a fuel source for peripheral tissues in mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Carnitine/analogs & derivatives , Lipids/blood , Liver/metabolism , Thermogenesis , Aging/physiology , Animals , Body Temperature , Carnitine/administration & dosage , Carnitine/blood , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Citric Acid Cycle , Cold Temperature , Fatty Acids/blood , Gene Expression Regulation , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 4/metabolism , Lipolysis , Liver/enzymology , Mice , Phenotype , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...