Dietary fructose feeds hepatic lipogenesis via microbiota-derived acetate

Steven Zhao, Cholsoon Jang, Joyce Liu, Kahealani Uehara, Michael Gilbert, Luke Izzo, Xianfeng Zeng, Sophie Trefely, Sully Fernandez, Alessandro Carrer, Katelyn D Miller, Zachary T Schug, Nathaniel W Snyder, Terence P Gade, Paul M Titchenell, Joshua D Rabinowitz, Kathryn E Wellen, Steven Zhao, Cholsoon Jang, Joyce Liu, Kahealani Uehara, Michael Gilbert, Luke Izzo, Xianfeng Zeng, Sophie Trefely, Sully Fernandez, Alessandro Carrer, Katelyn D Miller, Zachary T Schug, Nathaniel W Snyder, Terence P Gade, Paul M Titchenell, Joshua D Rabinowitz, Kathryn E Wellen

Abstract

Consumption of fructose has risen markedly in recent decades owing to the use of sucrose and high-fructose corn syrup in beverages and processed foods1, and this has contributed to increasing rates of obesity and non-alcoholic fatty liver disease2-4. Fructose intake triggers de novo lipogenesis in the liver4-6, in which carbon precursors of acetyl-CoA are converted into fatty acids. The ATP citrate lyase (ACLY) enzyme cleaves cytosolic citrate to generate acetyl-CoA, and is upregulated after consumption of carbohydrates7. Clinical trials are currently pursuing the inhibition of ACLY as a treatment for metabolic diseases8. However, the route from dietary fructose to hepatic acetyl-CoA and lipids remains unknown. Here, using in vivo isotope tracing, we show that liver-specific deletion of Acly in mice is unable to suppress fructose-induced lipogenesis. Dietary fructose is converted to acetate by the gut microbiota9, and this supplies lipogenic acetyl-CoA independently of ACLY10. Depletion of the microbiota or silencing of hepatic ACSS2, which generates acetyl-CoA from acetate, potently suppresses the conversion of bolus fructose into hepatic acetyl-CoA and fatty acids. When fructose is consumed more gradually to facilitate its absorption in the small intestine, both citrate cleavage in hepatocytes and microorganism-derived acetate contribute to lipogenesis. By contrast, the lipogenic transcriptional program is activated in response to fructose in a manner that is independent of acetyl-CoA metabolism. These data reveal a two-pronged mechanism that regulates hepatic lipogenesis, in which fructolysis within hepatocytes provides a signal to promote the expression of lipogenic genes, and the generation of microbial acetate feeds lipogenic pools of acetyl-CoA.

Conflict of interest statement

J.D.R. is a consultant to Pfizer and to Colorado Research Partners. All other authors declare no conflicts of interest.

Figures

ED Figure 1.. Hepatic ACLY deficiency minimally…
ED Figure 1.. Hepatic ACLY deficiency minimally impacts the response to dietary fructose.
a, Schematic of fructolysis and glycolysis feeding into de novo lipogenesis. F1P = fructose-1-phosphate, F-1,6-BP = fructose-1,6-bisphosphate, GA = glyceraldehyde, DHAP = dihydroxyacetone phosphate, G3P = glyceraldehyde-3-phosphate. b, Body weights of WT and LAKO mice on CD or HFrD for 18 weeks (n = WT-CD:13, LAKO-CD:5, WT-HFrD:14, LAKO-HFrD:5), data are mean ± SD. c, Weights of liver, posterior subcutaneous (sWAT) and perigonadal (pgWAT) adipose tissues in WT and LAKO mice on CD or HFrD for 18 weeks (n Liver/sWAT/pgWAT = WT – CD: 7/7/7, LAKO – CD: 2/5/5, WT – HFrD: 6/12/12, LAKO – HFrD: 3/5/5). d, Representative images of Periodic Acid Schiff (PAS) stain for glycogen and Trichrome (TC) histological stain for fibrosis in livers from WT or LAKO mice on HFrD. Scale bars = 100 μm. e, Triglyceride content in WT or LAKO mice on CD or HFrD for 18 weeks, n = (WT-CD: 4, LAKO-CD: 3, WT-HFrD: 4, LAKO-HFrD: 3), statistical significance determined using Welch’s t test. For panels c and e, bars represent mean.
Extended Data Figure 2.. Hepatic ACLY deficiency…
Extended Data Figure 2.. Hepatic ACLY deficiency results in modest metabolic alterations on high fructose diet.
a, Volcano plot of hepatic metabolites in WT and LAKO mice on CD or HFrD for 4 weeks, pink dots indicate significant hits as determined by a fold-change threshold of 2 and P-value threshold of 0.1, assuming equal variance. b, Principle component analysis of log-transformed data in Supplementary Table 1, each dot represents a unique sample, 95% CI shown in corresponding color. c, Relative metabolite abundance, normalized to WT-CD group, p values determined using Welch’s t test, n = (WT-CD:5, LAKO-CD: 3, WT-HFrD: 5, LAKO-HFrD: 4). Bars represent mean.
Extended Data Figure 3.. High fructose diet…
Extended Data Figure 3.. High fructose diet alters hepatic lipid metabolism.
a, Hierarchical clustering of relative hepatic triglyceride abundance in WT or LAKO mice on CD or HFrD for 4 weeks, clustering performed using one minus pearson correlation and average linkage. b, Relative abundance of hepatic triglycerides composed of 16:0 to 18:1 fatty acids, subset of data in panel a. c, Principle component analysis of log-transformed data in Supplementary Table 2, each dot represents a unique sample, 95% CI shown in corresponding color.
Extended Data Figure 4.. Fructose induces steatosis…
Extended Data Figure 4.. Fructose induces steatosis and contributes substantially to newly synthesized fatty acids in the liver independently of ACLY.
a, Schematic of experimental design of drinking water study. b, Daily consumption of unsweetened (H2O) or 15% fructose + 15% glucose sweetened (Fruc:Gluc) water per mouse, each dot represents a repeat measurement (n = H2O: 6, Fruc:Gluc: 7), statistical significance determined using Welch’s t test. c, Weight gain of WT or LAKO mice given H2O or Fruc:Gluc water for 4 weeks (n = WT – H2O: 4, LAKO – H2O: 4, WT – Fruc:Gluc: 8, LAKO Fruc:Gluc: 6), p value indicated comparing all H2O vs. Fruc:Gluc mice determined by Welch’s t test. d, Representative H&E and Oil Red O histological stains of livers from mice in panel c. Scale bars = 100 μm. e, Experimental design for data shown in Figure 1c. f, Isotopologue distribution of labeled serum saponified fatty acids shown in Figure 1c. For all panels, data are mean ± SD.
Extended Figure Data 5.. Fructose signals the…
Extended Figure Data 5.. Fructose signals the use of acetate for de novo lipogenesis.
a, mRNA expression of ChREBP and its target genes in livers of WT or LAKO mice fed CD or HFrD (n = 4 mice/group), p values for WT-CD vs. WT-HFrD (blue text) and LAKO-CD vs. LAKO-HFrD (purple text) determined using two-sided t tests with Holm-Sidak method for multiple comparisons. b, mRNA expression of lipogenic genes in livers of WT or LAKO mice given H2O or Fruc:Gluc water for 4 weeks (n = 4/group), statistical comparisons WT-H2O vs. WT-Fruc:Gluc, p values for WT – H2O vs. WT – Fruc:Gluc (blue font) and LAKO – H2O vs. LAKO Fruc:Gluc (purple font) determined using two-sided t test with Holm-Sidak method for multiple comparisons. c, Immunoblots of lipogenic enzymes in livers of WT or LAKO mice given H2O or Fruc:Gluc water for 4 weeks, each lane represents an individual mouse. d, Immunohistochemistry staining against ACLY in WT or LAKO livers on H2O or Fruc:Gluc water for 4 weeks. Yellow boxes approximate location of 20X panels. Scale bars = 100 μm for 10X, 50 μm for 20X. e, H3K27ac ChIP-qPCR of livers from WT mice provided either water for 24 hours followed by an oral gavage of saline, or Fruc:Gluc water for 24 hours, followed by an oral gavage of 2.0 g/kg glucose and 2.0 g/kg fructose (Mlxipl: n = 3, Acss2: n = 3, Pklr: n = 4), livers harvested 90 minutes after gavage. p1 and p2 are two different primer sets. f, ChIP-seq tracks of Mlxipl, Pklr, Acss2 genomic loci, red bars indicate genomic regions used to design ChIP-qPCR primers. For panels a-b and e-f, bars represent mean.
Extended Data Figure 6.. Depletion of microbiome…
Extended Data Figure 6.. Depletion of microbiome blocks substrate contribution, but not signaling component, of de novo lipogenesis following fructose consumption.
a, Experimental set-up for antibiotic depletion of the microbiome followed by 13C-fructose tracing into DNL. b, Representative images of cecums from a saline and antibiotic treated mouse. c, Relative abundance of bacterial abundance in cecal contents from mice treated with saline (n = 9) or antibiotics (n = 9) as determined by 16s RT-qPCR to a reference standard of E. coli DNA. P value determined using Welch’s t test. d, Heat map of microbial metabolite abundance in the portal blood, collected 1 hour after gavage. e, Relative abundance of portal blood 13C-fructose in WT – saline (n = 7) vs. WT – Antibiotics (n = 7) and LAKO – saline (n = 4) vs. LAKO – antibiotics (n = 4) following gavage and f, % total labeled carbons in portal blood glucose, p values determined using Welch’s t test. g, mRNA expression of ChREBPβ, Acss2, and Fasn in liver collected 1 hour after gavage (n = 4/group), p values determined using two-sided t tests with Holm-Sidak method for multiple comparisons. For c, e-g, bars represent mean.
Extended Data Figure 7.. Bolus fructose is…
Extended Data Figure 7.. Bolus fructose is converted into acetate in a microbiome-dependent manner.
a, TIC of labeled liver F1P, pyruvate, and acetyl-CoA, concentrations (μM) of portal blood labeled acetate, and % labeling of liver saponified 16:0 and 18:0 in saline- or antibiotic-treated WT mice gavaged with 2.0 g/kg 13C-fructose + 2.0 g/kg unlabeled glucose, n = 3 mice/time point. Data for saline-treated mice is also shown in Figure 2d. b, Isotopologue distribution of serum saponified fatty acids, collected 6 hours after gavage, data are mean ± SD, n = (WT-Saline: 8, LAKO-Saline: 4, WT-Antibiotics: 8, LAKO-Antibiotics: 4).
Extended Data Figure 8.. Bolus fructose-dependent DNL…
Extended Data Figure 8.. Bolus fructose-dependent DNL requires microbial acetate and hepatic ACSS2.
a, Concentrations (μM) of portal blood labeled acetate, propionate, and butyrate, n = (WT-Saline: 8, LAKO-Saline: 4, WT-Antibiotics: 8, LAKO-Antibiotics: 4). b, Abundance of cecal labeled acetate, propionate, and butyrate in WT mice, n = 3 mice/timepoint, except for saline-180 n = 2 mice. c, Heat map of hepatic triglyceride abundance in livers of saline- or antibiotic-treated mice following an oral gavage of 2.0 g/kg fructose + 2.0 g/kg glucose. d, Concentrations of portal and systemic blood acetate following gavage, each data point represents an individual mouse sacrificed at indicated time, p value determined using two-sided t tests with Holm-Sidak method for multiple comparisons. e, Weight gain in WT and LAKO mice 1 week following tail-vein injection with AAV8-GFP or AAV8-shAcss2, p value determined using Welch’s t test. f, Liver weight as % of body weight of WT and LAKO mice 1 week following tail-vein injection with AAV8-GFP or AAV8-shAcss2. g, Western blot of liver lysates from WT and LAKO mice 1 week following tail-vein injection with AAV8-GFP or AAV8-shAcss2.
Extended Data Figure 9.. Gradual fructose consumption…
Extended Data Figure 9.. Gradual fructose consumption promotes greater acetate usage in LAKO mice.
a, Experimental set-up for 13C-acetate tracing into DNL prior to and after gradual fructose administration. b, Western blot of ACLY, ACSS2, and S6 in liver lysates from WT and LAKO mice after 1 day or 14 days of Fruc:Gluc water. c, Representative H&E stains of livers from WT and LAKO mice provided Fruc:Gluc water for 2 weeks. Scale bars = 100 μm d, Relative abundance of acetate, propionate, and butyrate in the cecal contents of WT and LAKO mice treated with saline or antibiotics for 1 week (n = 4 mice/group). p values determined using Welch’s t test.
Extended Data Figure 10.. Fructose provides signal…
Extended Data Figure 10.. Fructose provides signal and substrate to promote hepatic de novo lipogenesis.
a, Proposed model of bolus fructose-induced hepatic DNL. Fructose catabolism in hepatocytes acts as a signal to induce DNL genes including ACSS2, while fructose metabolism by the gut microbiome provides acetate as a substrate to feed DNL, mediated by ACSS2. b, Proposed model of gradual fructose-induced hepatic DNL. Like the bolus model, fructose catabolism in hepatocytes acts as a signal to induce DNL genes. Hepatic fructose and glucose (made from fructose by the small intestine) catabolism provide citrate as a substrate to feed DNL, mediated by ACLY. Metabolism of fibers and other dietary components by the gut microbiome provides acetate as a substrate to feed DNL, following its conversion to acetyl-CoA by hepatic ACSS2. Created with BioRender.com.
Figure 1.. Fructose-dependent fatty acid synthesis is…
Figure 1.. Fructose-dependent fatty acid synthesis is ACLY-independent.
a, Representative H&E and Oil Red O histological stains of livers of WT or LAKO mice on chow (CD) or high fructose diet (HFrD) from two independent experiments of n = 4 WT and LAKO mice/diet (4 weeks) and n = 13 WT mice/diet and n = 6 LAKO mice/diet (18 weeks). Scale bars = 100 μm. b, Relative deuterium incorporation in palmitic acid (16:0) and stearic acid (18:0) after 24 hour D2O labeling of mice, D2O set to 1 and compared to D2O Fruc:Gluc within each genotype, significance determined by two-sided t tests. c, % total labeled carbons in serum saponified fatty acids from mice gavaged with 1:1 fructose:glucose, 13C-labeled substrate indicated, bars represent mean. d, Immunoblots of lipogenic enzymes in livers of WT or LAKO mice fed CD or HFrD for 4 weeks.
Figure 2.. Lipogenic acetyl-CoA is preferentially produced…
Figure 2.. Lipogenic acetyl-CoA is preferentially produced from acetate in hepatocytes.
a, Pathways for lipogenic acetyl-CoA production from fructose, glucose, or acetate. b-c, % total labeled carbons in fructolytic intermediates (b) and acetyl-CoA, malonyl-CoA, or HMG-CoA (c) in primary hepatocytes incubated for 6 hours with 25mM fructose + 1mM acetate, 13C-labeled substrate indicated, bars represent mean, n = 3. d, Total ion counts (TIC) of labeled liver F1P, pyruvate, and acetyl-CoA, concentrations (μM) of portal blood labeled acetate, and % labeling of liver 16:0 and 18:0 in saline-treated WT mice gavaged with 1:1 13C-fructose:glucose, data are mean ± SEM, n = 3 mice/time point.
Figure 3.. Metabolism of bolus fructose by…
Figure 3.. Metabolism of bolus fructose by the microbiome feeds hepatic lipogenesis.
a, Area under curve (AUC0–240 min) of labeled hepatic F1P, pyruvate, acetyl-CoA, palmitate and portal blood acetate, data are mean ± SEM. See Extended Data Figure 8a for curves. b, % total labeled carbons in serum saponified fatty acids from saline- or antibiotic-treated LAKO mice gavaged with 13C-fructose:glucose, significance determined using two-sided t tests. c, % total labeled carbons in serum saponified fatty acids from saline- or antibiotic-treated LAKO mice gavaged with 1:1 fructose:glucose + 0.5 g/kg acetate, 13C-labeled substrate indicated, significance determined by two-sided t test. d, % total labeled carbons in serum saponified fatty acids from WT and LAKO mice 1 week after injection with AAV-GFP or AAV-shAcss2, significance determined by two-sided t test. For b-c, bars represent mean.
Figure 4.. Gradual fructose consumption promotes hepatic…
Figure 4.. Gradual fructose consumption promotes hepatic lipogenesis from ACLY- and ACSS2-derived acetyl-CoA.
a, Experimental design for gradual fructose consumption. b, % total labeled carbons from 13C-fructose or glucose in hepatic saponified fatty acids, WT vs. LAKO. c, % total labeled hydrogens from D2O in hepatic saponified fatty acids. d, % total labeled carbons from 13C-acetate in serum saponified fatty acids, see Extended Data Fig. 10a for experimental design. e, total % labeled hydrogens from D2O in hepatic saponified fatty acids in WT and LAKO mice following 1 week treatment with saline or antibiotics (abx). f, mRNA expression of ChREBP and lipogenic genes in livers of mice in (e). g, total % hydrogens labeled in hepatic saponified fatty acids in WT and LAKO mice 1 week after injection with AAV-GFP or AAV-shAcss2. For b-g, bars represent mean, b-e,g, significance determined using 2-way ANOVA with Tukey’s test for multiple comparisons.

References

    1. Bray GA, Nielsen SJ & Popkin BM Consumption of high-fructose corn syrup in beverages may play a role in the epidemic of obesity. Am. J. Clin. Nutr. 79, 537–43 (2004).
    1. Jensen T et al. Fructose and sugar: A major mediator of non-alcoholic fatty liver disease. J. Hepatol. 68, 1063–1075 (2018).
    1. Hannou SA, Haslam DE, McKeown NM & Herman MA Fructose metabolism and metabolic disease. J. Clin. Invest. 128, 545–555 (2018).
    1. Softic S, Cohen DE & Kahn CR Role of Dietary Fructose and Hepatic De Novo Lipogenesis in Fatty Liver Disease. Dig. Dis. Sci. 61, 1282–1293 (2016).
    1. Lambert JE, Ramos-Roman MA, Browning JD & Parks EJ Increased de novo lipogenesis is a distinct characteristic of individuals with nonalcoholic fatty liver disease. Gastroenterology 146, 726–735 (2014).
    1. Donnelly KL et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Invest. 115, 1343–51 (2005).
    1. Fukuda H, Katsurada A & Iritani N Effects of nutrients and hormones on gene expression of ATP citrate-lyase in rat liver. Eur. J. Biochem. 209, 217–22 (1992).
    1. Pinkosky SL, Groot PHE, Lalwani ND & Steinberg GR Targeting ATP-Citrate Lyase in Hyperlipidemia and Metabolic Disorders. Trends Mol. Med. 23, 1047–1063 (2017).
    1. Jang C et al. The Small Intestine Converts Dietary Fructose into Glucose and Organic Acids. Cell Metab. 27, 351–361.e3 (2018).
    1. Zhao S et al. ATP-Citrate Lyase Controls a Glucose-to-Acetate Metabolic Switch. Cell Rep. 17, 1037–1052 (2016).
    1. Beigneux AP et al. ATP-citrate lyase deficiency in the mouse. J. Biol. Chem. 279, 9557–64 (2004).
    1. Herman MA & Samuel VT The Sweet Path to Metabolic Demise: Fructose and Lipid Synthesis. Trends Endocrinol. Metab. 27, 719–730 (2016).
    1. Uyeda K & Repa JJ Carbohydrate response element binding protein, ChREBP, a transcription factor coupling hepatic glucose utilization and lipid synthesis. Cell Metab. 4, 107–110 (2006).
    1. Herman MA et al. A novel ChREBP isoform in adipose tissue regulates systemic glucose metabolism. Nature 484, 333–8 (2012).
    1. Iizuka K The role of carbohydrate response element binding protein in intestinal and hepatic fructose metabolism. Nutrients 9, 1–12 (2017).
    1. Poungvarin N et al. Genome-Wide Analysis of ChREBP Binding Sites on Male Mouse Liver and White Adipose Chromatin. Endocrinology 156, 1982–94 (2015).
    1. Luong A, Hannah VC, Brown MS & Goldstein JL Molecular characterization of human acetyl-CoA synthetase, an enzyme regulated by sterol regulatory element-binding proteins. J. Biol. Chem. 275, 26458–26466 (2000).
    1. Ikeda Y et al. Transcriptional Regulation of the Murine Acetyl-CoA Synthetase 1 Gene through Multiple Clustered Binding Sites for Sterol Regulatory Element-binding Proteins and a Single Neighboring Site for Sp1. J. Biol. Chem. 276, 34259–34269 (2001).
    1. Softic S et al. Divergent effects of glucose and fructose on hepatic lipogenesis and insulin signaling. J. Clin. Invest. 127, 4059–4074 (2017).
    1. Liu X et al. Acetate Production from Glucose and Coupling to Mitochondrial Metabolism in Mammals. Cell 175, 502–513.e13 (2018).
    1. Bulusu V et al. Acetate Recapturing by Nuclear Acetyl-CoA Synthetase 2 Prevents Loss of Histone Acetylation during Oxygen and Serum Limitation. Cell Rep. 18, 647–658 (2017).
    1. Lu M et al. ACOT12-Dependent Alteration of Acetyl-CoA Drives Hepatocellular Carcinoma Metastasis by Epigenetic Induction of Epithelial-Mesenchymal Transition. Cell Metab. 1–15 (2019). doi:10.1016/j.cmet.2018.12.019
    1. Ter Horst KW & Serlie MJ Fructose consumption, lipogenesis, and non-alcoholic fatty liver disease. Nutrients 9, 1–20 (2017).
    1. Bergheim I et al. Antibiotics protect against fructose-induced hepatic lipid accumulation in mice: Role of endotoxin. J. Hepatol. 48, 983–992 (2008).
    1. Kaden-Volynets V et al. Lack of liver steatosis in germ-free mice following hypercaloric diets. Eur. J. Nutr. 0, 1–13 (2018).
    1. Mews P et al. Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory. Nature 546, 381–386 (2017).
    1. Li MV et al. Glucose-6-phosphate mediates activation of the carbohydrate responsive binding protein (ChREBP). Biochem. Biophys. Res. Commun. 395, 395–400 (2010).
    1. Lanaspa MA et al. Ketohexokinase C blockade ameliorates fructose-induced metabolic dysfunction in fructose-sensitive mice. J. Clin. Invest. 128, 2226–2238 (2018).
    1. Ishimoto T et al. Opposing effects of fructokinase C and A isoforms on fructose-induced metabolic syndrome in mice. Proc. Natl. Acad. Sci. U. S. A. 109, 4320–5 (2012).
    1. Parks EJ, Skokan LE, Timlin MT & Dingfelder CS Dietary Sugars Stimulate Fatty Acid Synthesis in Adults. J. Nutr. 1039–1046 (2008). doi:10.1016/j.bbi.2008.05.010
    1. den Besten G et al. Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids. Am. J. Physiol. - Gastrointest. Liver Physiol. 305, 900–910 (2013).
    1. Donohoe DR et al. The Warburg Effect Dictates the Mechanism of Butyrate-Mediated Histone Acetylation and Cell Proliferation. Mol. Cell 48, 612–626 (2012).
    1. Zagelbaum NK, Yandrapalli S, Nabors C & Frishman WH Bempedoic Acid (ETC-1002): ATP Citrate Lyase Inhibitor: Review of a First-in-Class Medication with Potential Benefit in Statin-Refractory Cases. Cardiol. Rev. 27, 49–56 (2018).
    1. Guo L et al. Enhanced acetylation of ATP-citrate lyase promotes the progression of nonalcoholic fatty liver disease. J. Biol. Chem. 294, 11805–11816 (2019).
    1. Wang Q et al. Abrogation of hepatic ATP-citrate lyase protects against fatty liver and ameliorates hyperglycemia in leptin receptor-deficient mice. Hepatology 49, 1166–75 (2009).
    1. Wang Q et al. Deficiency in hepatic ATP-citrate lyase affects VLDL-triglyceride mobilization and liver fatty acid composition in mice. J. Lipid Res. 51, 2516–26 (2010).
Method References
    1. Postic C et al. Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic b cell-specific gene knock-outs using Cre recombinase. J. Biol. Chem. 274, 305–315 (1999).
    1. Nadkarni MA, Martin FE, Jacques NA & Hunter N Determination of bacterial load by real-time PCR using a broad-range (universal) probe and primers set. Microbiology 148, 257–266 (2002).
    1. Guan D et al. Diet-Induced Circadian Enhancer Remodeling Synchronizes Opposing Hepatic Lipid Metabolic Processes. Cell 174, 831–842.e12 (2018).
    1. Su X, Lu W & Rabinowitz JD Metabolite Spectral Accuracy on Orbitraps. Anal. Chem. 89, 5940–5948 (2017).
    1. Titchenell PM, Chu Q, Monks BR & Birnbaum MJ Hepatic insulin signalling is dispensable for suppression of glucose output by insulin in vivo. Nat. Commun. 6, 1–9 (2015).
    1. Iroz A et al. A Specific ChREBP and PPARα Cross-Talk Is Required for the Glucose-Mediated FGF21 Response. Cell Rep. 21, 403–416 (2017).
    1. Frey AJ et al. LC-quadrupole/Orbitrap high-resolution mass spectrometry enables stable isotope-resolved simultaneous quantification and 13C-isotopic labeling of acyl-coenzyme A thioesters. Anal. Bioanal. Chem. 408, 3651–3658 (2016).
    1. Trefely S, Ashwell P & Snyder NW FluxFix: automatic isotopologue normalization for metabolic tracer analysis. BMC Bioinformatics 17, 485 (2016).
    1. Lee JV et al. Acetyl-CoA promotes glioblastoma cell adhesion and migration through Ca2+-NFAT signaling. Genes Dev. 32, (2018).
    1. Chong J et al. MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis. Nucleic Acids Res. 46, W486–W494 (2018).
    1. Neinast MD et al. Quantitative Analysis of the Whole-Body Metabolic Fate of Branched-Chain Amino Acids. Cell Metab. 1–13 (2018). doi:10.1016/j.cmet.2018.10.013

Source: PubMed

3
구독하다