Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Mol Cell ; 82(2): 447-462.e6, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34856123

ABSTRACT

Quantitative subcellular metabolomic measurements can explain the roles of metabolites in cellular processes but are subject to multiple confounding factors. We developed stable isotope labeling of essential nutrients in cell culture-subcellular fractionation (SILEC-SF), which uses isotope-labeled internal standard controls that are present throughout fractionation and processing to quantify acyl-coenzyme A (acyl-CoA) thioesters in subcellular compartments by liquid chromatography-mass spectrometry. We tested SILEC-SF in a range of sample types and examined the compartmentalized responses to oxygen tension, cellular differentiation, and nutrient availability. Application of SILEC-SF to the challenging analysis of the nuclear compartment revealed a nuclear acyl-CoA profile distinct from that of the cytosol, with notable nuclear enrichment of propionyl-CoA. Using isotope tracing, we identified the branched chain amino acid isoleucine as a major metabolic source of nuclear propionyl-CoA and histone propionylation, thus revealing a new mechanism of crosstalk between metabolism and the epigenome.


Subject(s)
Acyl Coenzyme A/metabolism , Cell Compartmentation , Cell Nucleus/metabolism , Energy Metabolism , Histones/metabolism , Metabolomics , Protein Processing, Post-Translational , Animals , Cell Differentiation , Chromatography, Liquid , Cytosol/metabolism , Epigenesis, Genetic , Hep G2 Cells , Humans , Isoleucine , Metabolome , Mice , Mitochondria/metabolism , Oxygen/metabolism , Spectrometry, Mass, Electrospray Ionization
2.
Free Radic Biol Med ; 174: 264-271, 2021 10.
Article in English | MEDLINE | ID: mdl-34371153

ABSTRACT

Oxidized phospholipids (OxPLs) containing enzymatically or non-enzymatically oxidized fatty acids (oxylipins) are increasingly recognized as lipid mediators involved in pathogenesis of diseases. Further understanding of structure-activity relationship and molecular mechanisms activated by OxPLs is hampered by the complexity of synthesis of individual molecular species. Although dozens of individual free oxylipins are commercially available, their attachment to the phospholipid scaffold requires relatively harsh conditions during activation of carboxy-group, which may lead to decomposition of unstable oxylipins. Furthermore, additional protection-deprotection steps are required for oxylipins containing hydroxy-groups. In this work we describe synthesis of OxPLs containing oxylipins bound at the sn-2-position via an amide-bond that is characteristic of sphingophospholipids. Activation of oxylipins and attachment to the phospholipid scaffold are performed under mild conditions and characterized by high yield. Hydroxy-groups of oxylipins do not interfere with reactions and therefore no protection/deprotection steps are needed. In order to prevent oxylipin migration, a fatty acid residue at the sn-1 was bound through an alkyl bond, which is a common bond present in a large proportion of naturally occurring phospholipids. An additional advantage of combining alkyl and amide bonds in a single phospholipid molecule is that both types of bonds are phospholipase A1/A2-resistant, which may be expected to improve biological stability of OxPLs and thus simplify analysis of their effects. As proof of principle, several alkyl-amide oxidized phosphatidylcholines (OxPCs) containing either linear or prostane ring oxylipins have been synthesized. Importantly, we show here that alkyl-amide-OxPCs demonstrated biological activities similar to those of di-acyl-OxPCs. Alkyl-amide-OxPCs inhibited pro-inflammatory action of LPS and increased endothelial cellular barrier in vitro and in mouse models. The effects of alkyl-amide and di-acyl-OxPCs developed in a similar range of concentrations. We hypothesize that alkyl-amide-OxPLs may become a useful tool for deeper analysis of the structure-activity relationship of OxPLs.


Subject(s)
Endotoxins , Phospholipids , Amides , Animals , Mice , Oxidation-Reduction , Phosphatidylcholines
3.
Cancer Metab ; 8(1): 26, 2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33292758

ABSTRACT

BACKGROUND: Aspartate biosynthesis and its delivery to the cytosol can be crucial for tumor growth in vivo. However, the impact of intracellular aspartate levels on metastasis has not been studied. We previously described that loss-of-aspartate glutamate carrier 1 (SLC25A12 or AGC1), an important component of the malate-aspartate shuttle, impairs cytosolic aspartate levels, NAD+/NADH ratio, mitochondrial respiration, and tumor growth. Here, we report the impact of AGC1-knockdown on metastasis. RESULTS: Low AGC1 expression correlates with worse patient prognosis in many cancers. AGC1-knockdown in mouse lung carcinoma and melanoma cell lines leads to increased pulmonary metastasis following subcutaneous or intravenous injections, respectively. On the other hand, conventional in vitro metastasis assays show no indication of increased metastasis capacity of AGC1-knockdown cells. CONCLUSION: This study highlights that certain branches of metabolism impact tumor growth and tumor metastasis differently. In addition, it also argues that commonly known metastasis indicators, including EMT genes, cell migration, or colony formation, do not always reflect metastatic capacity in vivo.

4.
Mol Metab ; 30: 61-71, 2019 12.
Article in English | MEDLINE | ID: mdl-31767181

ABSTRACT

OBJECTIVE: The dynamic regulation of metabolic pathways can be monitored by stable isotope tracing. Yet, many metabolites are part of distinct processes within different subcellular compartments. Standard isotope tracing experiments relying on analyses in whole cells may not accurately reflect compartmentalized metabolic processes. Analysis of compartmentalized metabolism and the dynamic interplay between compartments can potentially be achieved by stable isotope tracing followed by subcellular fractionation. Although it is recognized that metabolism can take place during biochemical fractionation of cells, a clear understanding of how such post-harvest metabolism impacts the interpretation of subcellular isotope tracing data and methods to correct for this are lacking. We set out to directly assess artifactual metabolism, enabling us to develop and test strategies to correct for it. We apply these techniques to examine the compartment-specific metabolic kinetics of 13C-labeled substrates targeting central metabolic pathways. METHODS: We designed a stable isotope tracing strategy to interrogate post-harvest metabolic activity during subcellular fractionation using liquid chromatography-mass spectrometry (LC-MS). RESULTS: We show that post-harvest metabolic activity occurs rapidly (within seconds) upon cell harvest. With further characterization we reveal that this post-harvest metabolism is enzymatic and reflects the metabolic capacity of the sub-cellular compartment analyzed, but it is limited in the extent of its propagation into downstream metabolites in metabolic pathways. We also propose and test a post-labeling strategy to assess the amount of post-harvest metabolism occurring in an experiment and then to adjust data to account for this. We validate this approach for both mitochondrial and cytosolic metabolic analyses. CONCLUSIONS: Our data indicate that isotope tracing coupled with sub-cellular fractionation can reveal distinct and dynamic metabolic features of cellular compartments, and that confidence in such data can be improved by applying a post-labeling correction strategy. We examine compartmentalized metabolism of acetate and glutamine and show that acetyl-CoA is turned over rapidly in the cytosol and acts as a pacemaker of anabolic metabolism in this compartment.


Subject(s)
Metabolic Networks and Pathways/physiology , Metabolomics/methods , Subcellular Fractions/metabolism , Acetyl Coenzyme A/metabolism , Animals , Cell Compartmentation , Cell Line , Chromatography, Liquid/methods , Fibroblasts , Hep G2 Cells , Humans , Isotope Labeling/methods , Kinetics , Mass Spectrometry/methods , Mice
5.
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
6.
Mol Cell Oncol ; 6(5): e1536843, 2019.
Article in English | MEDLINE | ID: mdl-31528687

ABSTRACT

Cancer cells rely on glutamine to fuel mitochondria, however it remains unclear whether this is needed for bioenergetic or biosynthetic pathways. Our study suggests that an essential function of mitochondrial glutamine metabolism is to provide aspartate to the cytosol where it can be used for nucleotide and protein synthesis.

7.
Biochim Biophys Acta Mol Cell Res ; 1866(3): 337-348, 2019 03.
Article in English | MEDLINE | ID: mdl-30595160

ABSTRACT

The discovery of significant amounts of metabolically active brown adipose tissue (BAT) in adult humans renders it a promising target for anti-obesity therapies by inducing weight loss through increased energy expenditure. The components of the N-acetylaspartate (NAA) pathway are highly abundant in BAT. Aspartate N-acetyltransferase (Asp-NAT, encoded by Nat8l) synthesizes NAA from acetyl-CoA and aspartate and increases energy expenditure in brown adipocytes. However, the exact mechanism how the NAA pathway contributes to accelerated mobilization and oxidation of lipids and the physiological regulation of the NAA pathway remained elusive. Here, we demonstrate that the expression of NAA pathway genes corresponds to nutrient availability and specifically responds to changes in exogenous glucose. NAA is preferentially produced from glucose-derived acetyl-CoA and aspartate and its concentration increases during adipogenesis. Overexpression of Nat8l drains glucose-derived acetyl-CoA into the NAA pool at the expense of cellular lipids and certain amino acids. Mechanistically, we elucidated that a combined activation of neutral and lysosomal (acid) lipolysis is responsible for the increased lipid degradation. Specifically, translocation of the transcription factor EB to the nucleus activates the biosynthesis of autophagosomes and lysosomes. Lipid degradation within lysosomes accompanied by adipose triglyceride lipase-mediated lipolysis delivers fatty acids for the support of elevated mitochondrial respiration. Together, our data suggest a crucial role of the NAA pathway in energy metabolism and metabolic adaptation in BAT.


Subject(s)
Adipocytes, Brown/metabolism , Aspartic Acid/analogs & derivatives , Nutrients/metabolism , Acetyl Coenzyme A/metabolism , Acetyltransferases/metabolism , Adipocytes, Brown/physiology , Adipogenesis/genetics , Adipogenesis/physiology , Adipose Tissue, Brown/metabolism , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Aspartic Acid/physiology , Energy Metabolism/physiology , Fatty Acids/metabolism , Glucose/metabolism , Lipid Metabolism/physiology , Lipids/physiology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Oxidation-Reduction
8.
Cell Metab ; 28(5): 706-720.e6, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30122555

ABSTRACT

Mitochondrial function is important for aspartate biosynthesis in proliferating cells. Here, we show that mitochondrial aspartate export via the aspartate-glutamate carrier 1 (AGC1) supports cell proliferation and cellular redox homeostasis. Insufficient cytosolic aspartate delivery leads to cell death when TCA cycle carbon is reduced following glutamine withdrawal and/or glutaminase inhibition. Moreover, loss of AGC1 reduces allograft tumor growth that is further compromised by treatment with the glutaminase inhibitor CB-839. Together, these findings argue that mitochondrial aspartate export sustains cell survival in low-glutamine environments and AGC1 inhibition can synergize with glutaminase inhibition to limit tumor growth.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Antiporters/metabolism , Aspartic Acid/metabolism , Cell Survival , Cytosol/metabolism , Glutamine/metabolism , Animals , Cell Line , Cell Proliferation , Citric Acid Cycle , Female , Humans , Mice, Inbred C57BL , Mitochondria/metabolism , Neoplasms/metabolism
9.
Proc Natl Acad Sci U S A ; 115(30): 7819-7824, 2018 07 24.
Article in English | MEDLINE | ID: mdl-29915037

ABSTRACT

Insulin increases glucose uptake into adipose tissue and muscle by increasing trafficking of the glucose transporter Glut4. In cultured adipocytes, the exocytosis of Glut4 relies on activation of the small G protein RalA by insulin, via inhibition of its GTPase activating complex RalGAP. Here, we evaluate the role of RalA in glucose uptake in vivo with specific chemical inhibitors and by generation of mice with adipocyte-specific knockout of RalGAPB. RalA was profoundly activated in brown adipose tissue after feeding, and its inhibition prevented Glut4 exocytosis. RalGAPB knockout mice with diet-induced obesity were protected from the development of metabolic disease due to increased glucose uptake into brown fat. Thus, RalA plays a crucial role in glucose transport in adipose tissue in vivo.


Subject(s)
Adipose Tissue, Brown/metabolism , Glucose/metabolism , Homeostasis , ral GTP-Binding Proteins/metabolism , 3T3-L1 Cells , Adipose Tissue, Brown/pathology , Animals , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Deletion , Glucose/genetics , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Mice , Mice, Knockout , ral GTP-Binding Proteins/genetics
10.
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
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(4): 467-478, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29374543

ABSTRACT

Lysosomal acid lipase (LAL) is the only known enzyme, which hydrolyzes cholesteryl esters and triacylglycerols in lysosomes of multiple cells and tissues. Here, we explored the role of LAL in brown adipose tissue (BAT). LAL-deficient (Lal-/-) mice exhibit markedly reduced UCP1 expression in BAT, modified BAT morphology with accumulation of lysosomes, and mitochondrial dysfunction, consequently leading to regular hypothermic events in mice kept at room temperature. Cold exposure resulted in reduced lipid uptake into BAT, thereby aggravating dyslipidemia and causing life threatening hypothermia in Lal-/- mice. Linking LAL as a potential regulator of lipoprotein lipase activity, we found Angptl4 mRNA expression upregulated in BAT. Our data demonstrate that LAL is critical for shuttling fatty acids derived from circulating lipoproteins to BAT during cold exposure. We conclude that inhibited lysosomal lipid hydrolysis in BAT leads to impaired thermogenesis in Lal-/- mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Fatty Acids/metabolism , Sterol Esterase/metabolism , Thermogenesis , Acetyl Coenzyme A/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/ultrastructure , Animals , Autophagy , Body Temperature , Carnitine/analogs & derivatives , Carnitine/metabolism , Cold Temperature , Disease Progression , Dyslipidemias/metabolism , Dyslipidemias/pathology , Energy Metabolism , Glucose/metabolism , Hypothermia, Induced , Lipid Droplets/metabolism , Lipolysis , Male , Mice, Inbred C57BL , Muscles/metabolism , Oxidation-Reduction , Oxygen Consumption , Sterol Esterase/deficiency , Uncoupling Protein 1/metabolism
12.
Article in English | MEDLINE | ID: mdl-28979238

ABSTRACT

N-acetylaspartate (NAA) is a highly abundant brain metabolite. Aberrant NAA concentrations have been detected in many pathological conditions and although the function of NAA has been extensively investigated in the brain it is still controversial. Only recently, a role of NAA has been reported outside the brain. In brown adipocytes, which show high expression of the NAA-producing and the NAA-cleaving enzyme, the metabolism of NAA has been implicated in lipid synthesis and histone acetylation. Increased expression of N-acetyltransferase 8-like (Nat8l, the gene encoding the NAA synthesizing enzyme) induces de novo lipogenesis and the brown adipocyte phenotype. Accordingly silencing of aspartoacylase, the NAA-cleaving enzyme, reduced brown adipocyte differentiation mechanistically by decreasing histone acetylation and gene transcription. Notably, the expression of Nat8l and the amount of NAA were also shown to be increased in several tumors and inversely correlate with patients' survival. Additionally, Nat8l silencing reduced cell proliferation in tumor and non-tumor cells, while NAA supplementation could rescue it. However, the mechanism behind has not yet been clarified. It remains to be addressed whether NAA per se and/or its catabolism to acetate and aspartate, metabolites that have both been implicated in tumor growth, are valuable targets for future therapies.

13.
FASEB J ; 31(9): 4088-4103, 2017 09.
Article in English | MEDLINE | ID: mdl-28559441

ABSTRACT

Adipocyte plasma membrane-associated protein (APMAP) has been described as an adipogenic factor in 3T3-L1 cells with unknown biochemical function; we therefore aimed to investigate the physiologic function of APMAP in vivo We generated Apmap-knockout mice and challenged them with an obesogenic diet to investigate their metabolic phenotype. We identified a novel truncated adipocyte-specific isoform of APMAP in mice that is produced by alternative transcription. Mice lacking the full-length APMAP protein, the only isoform that is expressed in humans, have an improved metabolic phenotype upon diet-induced obesity, indicated by enhanced insulin sensitivity, preserved glucose tolerance, increased respiratory exchange ratio, decreased inflammatory marker gene expression, and reduced adipocyte size. At the molecular level, APMAP interacts with the extracellular collagen cross-linking matrix proteins lysyl oxidase-like 1 and 3. On a high-fat diet, the expression of lysyl oxidase-like 1 and 3 is strongly decreased in Apmap-knockout mice, paralleled by reduced expression of profibrotic collagens and total collagen content in epididymal white adipose tissue, indicating decreased fibrotic potential. Together, our data suggest that APMAP is a novel regulator of extracellular matrix components, and establish that APMAP is a potential target to mitigate obesity-associated insulin resistance.-Pessentheiner, A. R., Huber, K., Pelzmann, H. J., Prokesch, A., Radner, F. P. W., Wolinski, H., Lindroos-Christensen, J., Hoefler, G., Rülicke, T., Birner-Gruenberger, R., Bilban, M., Bogner-Strauss, J. G. APMAP interacts with lysyl oxidase-like proteins, and disruption of Apmap leads to beneficial visceral adipose tissue expansion.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Gene Expression Regulation/physiology , Intra-Abdominal Fat/metabolism , Membrane Glycoproteins/metabolism , Adipocytes/cytology , Adipocytes/physiology , Amino Acid Oxidoreductases/genetics , Animals , Cell Size , Diet, High-Fat , Down-Regulation , Humans , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Obesity , Protein Isoforms
14.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(3): 358-368, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28017862

ABSTRACT

The importance of peroxisomes for adipocyte function is poorly understood. Herein, we provide insights into the critical role of peroxin 16 (PEX16)-mediated peroxisome biogenesis in adipocyte development and lipid metabolism. Pex16 is highly expressed in adipose tissues and upregulated during adipogenesis of murine and human cells. We demonstrate that Pex16 is a target gene of the adipogenesis "master-regulator" PPARγ. Stable silencing of Pex16 in 3T3-L1 cells strongly reduced the number of peroxisomes while mitochondrial number was unaffected. Concomitantly, peroxisomal fatty acid (FA) oxidation was reduced, thereby causing accumulation of long- and very long-chain (polyunsaturated) FAs and reduction of odd-chain FAs. Further, Pex16-silencing decreased cellular oxygen consumption and increased FA release. Additionally, silencing of Pex16 impaired adipocyte differentiation, lipogenic and adipogenic marker gene expression, and cellular triglyceride stores. Addition of PPARγ agonist rosiglitazone and peroxisome-related lipid species to Pex16-silenced 3T3-L1 cells rescued adipogenesis. These data provide evidence that PEX16 is required for peroxisome biogenesis and highlights the relevance of peroxisomes for adipogenesis and adipocyte lipid metabolism.


Subject(s)
Adipocytes, White/metabolism , Homeostasis/physiology , Lipid Metabolism/physiology , Lipids/physiology , Membrane Proteins/metabolism , Peroxisomes/metabolism , 3T3-L1 Cells , Adipogenesis/physiology , Animals , COS Cells , Cell Differentiation/physiology , Cell Line , Chlorocebus aethiops , Fatty Acids/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Oxygen Consumption/physiology , PPAR gamma/metabolism , Up-Regulation/physiology
15.
FASEB J ; 31(2): 732-742, 2017 02.
Article in English | MEDLINE | ID: mdl-27811061

ABSTRACT

The ability to adapt cellular metabolism to nutrient availability is critical for survival. The liver plays a central role in the adaptation to starvation by switching from glucose-consuming processes and lipid synthesis to providing energy substrates like glucose to the organism. Here we report a previously unrecognized role of the tumor suppressor p53 in the physiologic adaptation to food withdrawal. We found that starvation robustly increases p53 protein in mouse liver. This induction was posttranscriptional and mediated by a hepatocyte-autonomous and AMP-activated protein kinase-dependent mechanism. p53 stabilization was required for the adaptive expression of genes involved in amino acid catabolism. Indeed, acute deletion of p53 in livers of adult mice impaired hepatic glycogen storage and induced steatosis. Upon food withdrawal, p53-deleted mice became hypoglycemic and showed defects in the starvation-associated utilization of hepatic amino acids. In summary, we provide novel evidence for a p53-dependent integration of acute changes of cellular energy status and the metabolic adaptation to starvation. Because of its tumor suppressor function, p53 stabilization by starvation could have implications for both metabolic and oncological diseases of the liver.-Prokesch, A., Graef, F. A., Madl, T., Kahlhofer, J., Heidenreich, S., Schumann, A., Moyschewitz, E., Pristoynik, P., Blaschitz, A., Knauer, M., Muenzner, M., Bogner-Strauss, J. G., Dohr, G., Schulz, T. J., Schupp, M. Liver p53 is stabilized upon starvation and required for amino acid catabolism and gluconeogenesis.


Subject(s)
Food Deprivation/physiology , Hepatocytes/physiology , Liver/metabolism , Tumor Suppressor Protein p53/metabolism , Adenylate Kinase/genetics , Adenylate Kinase/metabolism , Animals , Cells, Cultured , Fatty Liver/metabolism , Gene Deletion , Gene Expression Regulation , Gene Silencing , Glycogen/metabolism , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Transcriptome , Tumor Suppressor Protein p53/genetics
16.
Diabetes ; 64(4): 1120-30, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25315009

ABSTRACT

The current dogma is that obesity-associated hepatic inflammation is due to increased Kupffer cell (KC) activation. However, recruited hepatic macrophages (RHMs) were recently shown to represent a sizable liver macrophage population in the context of obesity. Therefore, we assessed whether KCs and RHMs, or both, represent the major liver inflammatory cell type in obesity. We used a combination of in vivo macrophage tracking methodologies and adoptive transfer techniques in which KCs and RHMs are differentially labeled with fluorescent markers. With these approaches, the inflammatory phenotype of these distinct macrophage populations was determined under lean and obese conditions. In vivo macrophage tracking revealed an approximately sixfold higher number of RHMs in obese mice than in lean mice, whereas the number of KCs was comparable. In addition, RHMs comprised smaller size and immature, monocyte-derived cells compared with KCs. Furthermore, RHMs from obese mice were more inflamed and expressed higher levels of tumor necrosis factor-α and interleukin-6 than RHMs from lean mice. A comparison of the MCP-1/C-C chemokine receptor type 2 (CCR2) chemokine system between the two cell types showed that the ligand (MCP-1) is more highly expressed in KCs than in RHMs, whereas CCR2 expression is approximately fivefold greater in RHMs. We conclude that KCs can participate in obesity-induced inflammation by causing the recruitment of RHMs, which are distinct from KCs and are not precursors to KCs. These RHMs then enhance the severity of obesity-induced inflammation and hepatic insulin resistance.


Subject(s)
Gluconeogenesis/physiology , Liver/metabolism , Macrophages/metabolism , Obesity/metabolism , Animals , Diet, High-Fat/adverse effects , Fatty Liver/metabolism , Fatty Liver/pathology , Interleukin-6/metabolism , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/pathology , Macrophages/pathology , Male , Mice , Mice, Obese , Obesity/etiology , Obesity/pathology , Receptors, CCR2/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Nat Med ; 20(8): 942-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24997608

ABSTRACT

It is well known that the ω-3 fatty acids (ω-3-FAs; also known as n-3 fatty acids) can exert potent anti-inflammatory effects. Commonly consumed as fish products, dietary supplements and pharmaceuticals, ω-3-FAs have a number of health benefits ascribed to them, including reduced plasma triglyceride levels, amelioration of atherosclerosis and increased insulin sensitivity. We reported that Gpr120 is the functional receptor for these fatty acids and that ω-3-FAs produce robust anti-inflammatory, insulin-sensitizing effects, both in vivo and in vitro, in a Gpr120-dependent manner. Indeed, genetic variants that predispose to obesity and diabetes have been described in the gene encoding GPR120 in humans (FFAR4). However, the amount of fish oils that would have to be consumed to sustain chronic agonism of Gpr120 is too high to be practical, and, thus, a high-affinity small-molecule Gpr120 agonist would be of potential clinical benefit. Accordingly, Gpr120 is a widely studied drug discovery target within the pharmaceutical industry. Gpr40 is another lipid-sensing G protein-coupled receptor, and it has been difficult to identify compounds with a high degree of selectivity for Gpr120 over Gpr40 (ref. 11). Here we report that a selective high-affinity, orally available, small-molecule Gpr120 agonist (cpdA) exerts potent anti-inflammatory effects on macrophages in vitro and in obese mice in vivo. Gpr120 agonist treatment of high-fat diet-fed obese mice causes improved glucose tolerance, decreased hyperinsulinemia, increased insulin sensitivity and decreased hepatic steatosis. This suggests that Gpr120 agonists could become new insulin-sensitizing drugs for the treatment of type 2 diabetes and other human insulin-resistant states in the future.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Fatty Acids, Omega-3/metabolism , Insulin Resistance/physiology , Receptors, G-Protein-Coupled/agonists , Animals , Arginase/biosynthesis , B-Lymphocytes, Regulatory/immunology , Base Sequence , Diabetes Mellitus, Type 2/genetics , Docosahexaenoic Acids/pharmacology , Fatty Liver/drug therapy , Hyperinsulinism/drug therapy , Inflammation , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Molecular Sequence Data , Nitric Oxide Synthase Type II/biosynthesis , Obesity/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , T-Lymphocytes, Regulatory/immunology
18.
BMC Genomics ; 14: 758, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24191950

ABSTRACT

BACKGROUND: Fasting induces specific molecular and metabolic adaptions in most organisms. In biomedical research fasting is used in metabolic studies to synchronize nutritional states of study subjects. Because there is a lack of standardization for this procedure, we need a deeper understanding of the dynamics and the molecular mechanisms in fasting. RESULTS: We investigated the dynamic changes of liver gene expression and serum parameters of mice at several time points during a 48 hour fasting experiment and then focused on the global gene expression changes in epididymal white adipose tissue (WAT) as well as on pathways common to WAT, liver, and skeletal muscle. This approach produced several intriguing insights: (i) rather than a sequential activation of biochemical pathways in fasted liver, as current knowledge dictates, our data indicates a concerted parallel response; (ii) this first characterization of the transcriptome signature of WAT of fasted mice reveals a remarkable activation of components of the transcription apparatus; (iii) most importantly, our bioinformatic analyses indicate p53 as central node in the regulation of fasting in major metabolic tissues; and (iv) forced expression of Ddit4, a fasting-regulated p53 target gene, is sufficient to augment lipolysis in cultured adipocytes. CONCLUSIONS: In summary, this combination of focused and global profiling approaches provides a comprehensive molecular characterization of the processes operating during fasting in mice and suggests a role for p53, and its downstream target Ddit4, as novel components in the transcriptional response to food deprivation.


Subject(s)
Transcription Factors/metabolism , Transcriptome , Tumor Suppressor Protein p53/metabolism , Adipose Tissue, White/metabolism , Animals , Cell Line , Food Deprivation , Gene Expression Profiling , Gluconeogenesis , Lipogenesis , Lipolysis , Liver/metabolism , Male , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Annotation , Muscle, Skeletal/metabolism , Oligonucleotide Array Sequence Analysis , Signal Transduction , Stress, Physiological , Transcription Factors/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Up-Regulation
19.
PLoS One ; 8(11): e79134, 2013.
Article in English | MEDLINE | ID: mdl-24236098

ABSTRACT

Our knowledge about adipocyte metabolism and development is steadily growing, yet many players are still undefined. Here, we show that α/ß-hydrolase domain containing protein 15 (Abhd15) is a direct and functional target gene of peroxisome proliferator-activated receptor gamma (PPARγ), the master regulator of adipogenesis. In line, Abhd15 is mainly expressed in brown and white adipose tissue and strongly upregulated during adipogenesis in various murine and human cell lines. Stable knockdown of Abhd15 in 3T3-L1 cells evokes a striking differentiation defect, as evidenced by low lipid accumulation and decreased expression of adipocyte marker genes. In preconfluent cells, knockdown of Abhd15 leads to impaired proliferation, which is caused by apoptosis, as we see an increased SubG1 peak, caspase 3/7 activity, and BAX protein expression as well as a reduction in anti-apoptotic BCL-2 protein. Furthermore, apoptosis-inducing amounts of palmitic acid evoke a massive increase of Abhd15 expression, proposing an apoptosis-protecting role for ABHD15. On the other hand, in mature adipocytes physiological (i.e. non-apoptotic) concentrations of palmitic acid down-regulate Abhd15 expression. Accordingly, we found that the expression of Abhd15 in adipose tissue is reduced in physiological situations with high free fatty acid levels, like high-fat diet, fasting, and aging as well as in genetically obese mice. Collectively, our results position ABHD15 as an essential component in the development of adipocytes as well as in apoptosis, thereby connecting two substantial factors in the regulation of adipocyte number and size. Together with its intricate regulation by free fatty acids, ABHD15 might be an intriguing new target in obesity and diabetes research.


Subject(s)
Apoptosis , Carboxylic Ester Hydrolases/genetics , Membrane Proteins/genetics , 3T3-L1 Cells , Adipogenesis , Animals , Carboxylic Ester Hydrolases/metabolism , Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation, Enzymologic , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , PPAR gamma/physiology
20.
J Biol Chem ; 288(50): 36040-51, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24155240

ABSTRACT

NAT8L (N-acetyltransferase 8-like) catalyzes the formation of N-acetylaspartate (NAA) from acetyl-CoA and aspartate. In the brain, NAA delivers the acetate moiety for synthesis of acetyl-CoA that is further used for fatty acid generation. However, its function in other tissues remained elusive. Here, we show for the first time that Nat8l is highly expressed in adipose tissues and murine and human adipogenic cell lines and is localized in the mitochondria of brown adipocytes. Stable overexpression of Nat8l in immortalized brown adipogenic cells strongly increases glucose incorporation into neutral lipids, accompanied by increased lipolysis, indicating an accelerated lipid turnover. Additionally, mitochondrial mass and number as well as oxygen consumption are elevated upon Nat8l overexpression. Concordantly, expression levels of brown marker genes, such as Prdm16, Cidea, Pgc1α, Pparα, and particularly UCP1, are markedly elevated in these cells. Treatment with a PPARα antagonist indicates that the increase in UCP1 expression and oxygen consumption is PPARα-dependent. Nat8l knockdown in brown adipocytes has no impact on cellular triglyceride content, lipogenesis, or oxygen consumption, but lipolysis and brown marker gene expression are increased; the latter is also observed in BAT of Nat8l-KO mice. Interestingly, the expression of ATP-citrate lyase is increased in Nat8l-silenced adipocytes and BAT of Nat8l-KO mice, indicating a compensatory mechanism to sustain the acetyl-CoA pool once Nat8l levels are reduced. Taken together, our data show that Nat8l impacts on the brown adipogenic phenotype and suggests the existence of the NAT8L-driven NAA metabolism as a novel pathway to provide cytosolic acetyl-CoA for lipid synthesis in adipocytes.


Subject(s)
Acetyltransferases/metabolism , Adipocytes, Brown/metabolism , Energy Metabolism , Lipid Metabolism , Acetyl Coenzyme A/metabolism , Acetyltransferases/deficiency , Acetyltransferases/genetics , Adipocytes, Brown/cytology , Adipocytes, Brown/enzymology , Adipogenesis , Animals , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Enzymologic , Gene Knockout Techniques , Gene Silencing , Humans , Ion Channels/metabolism , Kinetics , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Size , PPAR alpha/metabolism , Phenotype , Phosphoproteins/metabolism , Protein Kinases/genetics , Protein Transport , Uncoupling Protein 1 , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...