Personal model-assisted identification of NAD+ and glutathione metabolism as intervention target in NAFLD

Adil Mardinoglu, Elias Bjornson, Cheng Zhang, Martina Klevstig, Sanni Söderlund, Marcus Ståhlman, Martin Adiels, Antti Hakkarainen, Nina Lundbom, Murat Kilicarslan, Björn M Hallström, Jesper Lundbom, Bruno Vergès, Peter Hugh R Barrett, Gerald F Watts, Mireille J Serlie, Jens Nielsen, Mathias Uhlén, Ulf Smith, Hanns-Ulrich Marschall, Marja-Riitta Taskinen, Jan Boren, Adil Mardinoglu, Elias Bjornson, Cheng Zhang, Martina Klevstig, Sanni Söderlund, Marcus Ståhlman, Martin Adiels, Antti Hakkarainen, Nina Lundbom, Murat Kilicarslan, Björn M Hallström, Jesper Lundbom, Bruno Vergès, Peter Hugh R Barrett, Gerald F Watts, Mireille J Serlie, Jens Nielsen, Mathias Uhlén, Ulf Smith, Hanns-Ulrich Marschall, Marja-Riitta Taskinen, Jan Boren

Abstract

To elucidate the molecular mechanisms underlying non-alcoholic fatty liver disease (NAFLD), we recruited 86 subjects with varying degrees of hepatic steatosis (HS). We obtained experimental data on lipoprotein fluxes and used these individual measurements as personalized constraints of a hepatocyte genome-scale metabolic model to investigate metabolic differences in liver, taking into account its interactions with other tissues. Our systems level analysis predicted an altered demand for NAD+ and glutathione (GSH) in subjects with high HS Our analysis and metabolomic measurements showed that plasma levels of glycine, serine, and associated metabolites are negatively correlated with HS, suggesting that these GSH metabolism precursors might be limiting. Quantification of the hepatic expression levels of the associated enzymes further pointed to altered de novo GSH synthesis. To assess the effect of GSH and NAD+ repletion on the development of NAFLD, we added precursors for GSH and NAD+ biosynthesis to the Western diet and demonstrated that supplementation prevents HS in mice. In a proof-of-concept human study, we found improved liver function and decreased HS after supplementation with serine (a precursor to glycine) and hereby propose a strategy for NAFLD treatment.

Keywords: NAFLD; glutathione; personalized genome‐scale metabolic modeling; serine.

© 2017 The Authors. Published under the terms of the CC BY 4.0 license.

Figures

Figure 1. Generation of VLDL kinetics and…
Figure 1. Generation of VLDL kinetics and plasma metabolomics data
  1. To identify the metabolic changes in response to increased hepatic steatosis (HS), secretion rate of the VLDLs from the liver of subjects was measured and the plasma metabolite levels were detected in subjects with varying degrees of HS.

  2. BMI, insulin resistance (HOMA‐IR), plasma TGs, and ALT levels are significantly correlated with the independently measured liver fat.

  3. The subjects were categorized into two groups as high (n = 43) and low HS (n = 43). Body mass index (BMI), fasting plasma insulin (FPI), plasma triglycerides (TGs), and plasma alanine aminotransferase (ALT) levels are found to be significantly different between the two groups. Data are presented as means ± SD. *P < 0.05; Student's t‐test.

Figure 2. Personalized modeling of liver in…
Figure 2. Personalized modeling of liver in subjects with varying degrees of HS
  1. A

    Schematic illustration of how personalized genome‐scale metabolic modeling can be performed accounting the interactions between other tissues and red blood cells for the development of effective therapeutic approaches for non‐alcoholic fatty liver disease (NAFLD). Solid and dashed arrows show the outputs and inputs to the tissues, respectively.

  2. B, C

    The correlation between the predicted intracellular fluxes of the liver and hepatic steatosis (HS) is assessed and compared with the (B) apolipoprotein B (apoB) and (C) triglycerides (TG) content in the total VLDL production.

  3. D

    The fluxes carried by the reactions catalyzed by NNT, GSR, and GPXs are found to be the most correlated reactions with the HS (Dataset EV3). Green arrow indicates the significant correlation of the flux carried by the reactions and HS.

  4. E

    The net fat influx (NFI) is calculated as the differences in the uptake and secretion rates of FAs and its correlation with the intracellular fluxes are assessed.

Figure 3. Correlation of the HS and…
Figure 3. Correlation of the HS and plasma metabolomics data
  1. A, B

    Hepatic steatosis (HS) is measured by the magnetic resonance imaging, and the plasma level of ˜520 metabolites was detected by untargeted metabolomics profiling. (A) The correlation between the HS and the plasma metabolites and (B) the Pearson correlation between the significantly (P < 0.05) correlated metabolites are presented. Red and blue colors represent the positive and negative correlation of the HS and plasma metabolite levels, respectively. Metabolites that can be converted to glycine marked bold.

Figure 4. Identification of significantly changed metabolites…
Figure 4. Identification of significantly changed metabolites in subjects with high HS
The plasma level of ~520 metabolites was detected by untargeted metabolomics profiling and significantly (P < 0.05) changed metabolites are presented using volcano plot. Metabolites that can be converted to glycine marked bold.
Figure 5. Glycine is the limiting substrate…
Figure 5. Glycine is the limiting substrate in the synthesis of GSH in NAFLD
  1. A

    Glycine is found to be the limiting substrate for the synthesis of glutathione (GSH) in subjects with NAFLD. Decreased de novo synthesis of serine has also been reported in subjects with NASH. The decreased plasma level of glycine, serine as well as other associated metabolites betaine and N‐acetylglycine in subjects with high HS is confirmed with the metabolomics study. In order to confirm model‐based predictions, serine was supplemented to the subjects with high HS since serine‐derived carbon can be converted to GSH to satisfy the increased demand for GSH in NAFLD. Blue arrows indicated downregulation whereas the red arrow indicates upregulation of the reactions. Green arrows indicate the significant correlation of the flux carried by the reactions and HS. [c], cytoplasm; [m], mitochondria.

  2. B–E

    The mRNA expressions of the (B) nicotinamide nucleotide transhydrogenase (NNT), (C) glutathione reductase (GSR), (D) glutamate–cysteine ligase, catalytic subunit (GCLC), and (E) glutamate–cysteine ligase, modifier subunit (GCLM) are measured in the livers obtained from 12 morbidly obese subjects who had undergone bariatric surgery and seven healthy individuals (mean ± SD). *P < 0.05; Student's t‐test.

Figure 6. Supplementation of NAD + and…
Figure 6. Supplementation of NAD+ and GSH precursors prevent NAFLD
Ten mice were given the Western diet supplemented with NR (400 mg/kg/day) and serine (300 mg/kg/day) la gavage and NAC (1 g/l) in the drinking water, and ten mice were only given the Western diet for 14 days.
  1. A–E

    Hepatic lipids including (A) triglycerides (TG), (B) cholesterol esters (CE), (C) ceramides (CER), (D) sphingomyelin (SM), (E) phosphatidylethanolamine (PE) (normalized to phosphatidylcholine (PC)) are shown in treated (cocktail supplemented) (n = 10), and control (n = 10) mice (mean ± SEM).

  2. F

    Quantification of amino acids from the liver of the same mice before and after supplementation (mean ± SEM).

  3. G

    Analysis of the molecular species of TGs extracted from the livers of the mice. Results from the control group are expressed as 100%, and results from the treated group are expressed as % of the control group (mean ± SEM).

  4. H

    mRNA expression of the fatty acid synthase (Fasn) in the liver tissue of mice before and after the supplementation. Six subjects received one oral dose of L‐serine (200 mg/kg/day) for 14 days (mean ± SEM).

  5. I–L

    The human plasma (I) alanine aminotransferase (ALT), (J) aspartate aminotransferase (AST), (K) alkaline phosphatase (ALP), and (L) TGs levels are presented in each human subject involved in the study before and after the supplementation with serine.

Data information: (A–G) *P < 0.05, **P < 0.01, ***<0.001; Student's t‐test.Source data are available online for this figure.
Figure 7. Three‐step strategy for the treatment…
Figure 7. Three‐step strategy for the treatment of NAFLD and associated disorders
Based on our results, we postulate a potential treatment strategy for NAFLD patients based on increased oxidation of fat and increased synthesis of GSH. A cocktail can be supplemented to NAFLD patients to boost these metabolic processes and to decrease the hepatic lipid accumulation. NR can be supplemented to boost the oxidation of the fat in the mitochondria by generating NAD+. Serine and NAC can be included in the cocktail to boost the level of GSH which is required for preventing the accumulation of incomplete products of fatty acids oxidation. l‐carnitine can also be added to the cocktail to boost the fatty acid uptake into mitochondria.

References

    1. Abdelmalek MF, Angulo P, Jorgensen RA, Sylvestre PB, Lindor KD (2001) Betaine, a promising new agent for patients with nonalcoholic steatohepatitis: results of a pilot study. Am J Gastroenterol 96: 2711–2717
    1. Adiels M, Packard C, Caslake MJ, Stewart P, Soro A, Westerbacka J, Wennberg B, Olofsson SO, Taskinen MR, Boren J (2005) A new combined multicompartmental model for apolipoprotein B‐100 and triglyceride metabolism in VLDL subfractions. J Lipid Res 46: 58–67
    1. Adiels M, Taskinen MR, Packard C, Caslake MJ, Soro‐Paavonen A, Westerbacka J, Vehkavaara S, Hakkinen A, Olofsson SO, Yki‐Jarvinen H, Boren J (2006) Overproduction of large VLDL particles is driven by increased liver fat content in man. Diabetologia 49: 755–765
    1. Adiels M, Mardinoglu A, Taskinen MR, Boren J (2015) Kinetic studies to elucidate impaired metabolism of triglyceride‐rich lipoproteins in humans. Front Physiol 6: 342
    1. Agren R, Mardinoglu A, Asplund A, Kampf C, Uhlen M, Nielsen J (2014) Identification of anticancer drugs for hepatocellular carcinoma through personalized genome‐scale metabolic modeling. Mol Syst Biol 10: 721
    1. Ardilouze JL, Fielding BA, Currie JM, Frayn KN, Karpe F (2004) Nitric oxide and beta‐adrenergic stimulation are major regulators of preprandial and postprandial subcutaneous adipose tissue blood flow in humans. Circulation 109: 47–52
    1. Benga G, Ferdinand W (1995) Amino acid composition of rat and human liver microsomes in normal and pathological conditions. Biosci Rep 15: 111–116
    1. Bickerton AST, Roberts R, Fielding BA, Hodson L, Blaak EE, Wagenmakers AJM, Gilbert M, Karpe F, Frayn KN (2007) Preferential uptake of dietary fatty acids in adipose tissue and muscle in the postprandial period. Diabetes 56: 168–176
    1. Boren J, Taskinen MR, Adiels M (2012) Kinetic studies to investigate lipoprotein metabolism. J Intern Med 271: 166–173
    1. Bourre JM (2006) Effects of nutrients (in food) on the structure and function of the nervous system: update on dietary requirements for brain. Part 2: macronutrients. J Nutr Health Aging 10: 386–399
    1. Canto C, Houtkooper RH, Pirinen E, Youn DY, Oosterveer MH, Cen Y, Fernandez‐Marcos PJ, Yamamoto H, Andreux PA, Cettour‐Rose P, Gademann K, Rinsch C, Schoonjans K, Sauve AA, Auwerx J (2012) The NAD(+) precursor nicotinamide riboside enhances oxidative metabolism and protects against high‐fat diet‐induced obesity. Cell Metab 15: 838–847
    1. de Carvalho SC, Muniz MT, Siqueira MD, Siqueira ER, Gomes AV, Silva KA, Bezerra LC, D'Almeida V, de Oliveira CP, Pereira LM (2013) Plasmatic higher levels of homocysteine in non‐alcoholic fatty liver disease (NAFLD). Nutr J 12: 37
    1. Clark RV, Walker AC, O'Connor‐Semmes RL, Leonard MS, Miller RR, Stimpson SA, Turner SM, Ravussin E, Cefalu WT, Hellerstein MK, Evans WJ (2014) Total body skeletal muscle mass: estimation by creatine (methyl‐d3) dilution in humans. J Appl Physiol 116: 1605–1613
    1. Cuervo AM, Dice JF (1996) A receptor for the selective uptake and degradation of proteins by lysosomes. Science 273: 501–503
    1. Deurenberg P, Weststrate JA, Seidell JC (1991) Body‐mass index as a measure of body fatness – age‐specific and sex‐specific prediction formulas. Br J Nutr 65: 105–114
    1. Dumas ME, Kinross J, Nicholson JK (2014) Metabolic phenotyping and systems biology approaches to understanding metabolic syndrome and fatty liver disease. Gastroenterology 146: 46–62
    1. Dyson JK, Anstee QM, McPherson S (2014) Non‐alcoholic fatty liver disease: a practical approach to treatment. Frontline Gastroenterol 5: 277–286
    1. Fernandez‐Checa JC, Kaplowitz N (2005) Hepatic mitochondrial glutathione: transport and role in disease and toxicity. Toxicol Appl Pharmacol 204: 263–273
    1. Folger O, Jerby L, Frezza C, Gottlieb E, Ruppin E, Shlomi T (2011) Predicting selective drug targets in cancer through metabolic networks. Mol Syst Biol 7: 501
    1. Foster DW (2012) Malonyl‐CoA: the regulator of fatty acid synthesis and oxidation. J Clin Investig 122: 1958–1959
    1. Frayn KN, Karpe F (2014) Regulation of human subcutaneous adipose tissue blood flow. Int J Obes 38: 1019–1026
    1. Frezza C, Zheng L, Folger O, Rajagopalan KN, MacKenzie ED, Jerby L, Micaroni M, Chaneton B, Adam J, Hedley A, Kalna G, Tomlinson IP, Pollard PJ, Watson DG, Deberardinis RJ, Shlomi T, Ruppin E, Gottlieb E (2011) Haem oxygenase is synthetically lethal with the tumour suppressor fumarate hydratase. Nature 477: 225–228
    1. Garcia‐Canaveras JC, Donato MT, Castell JV, Lahoz A (2011) A comprehensive untargeted metabonomic analysis of human steatotic liver tissue by RP and HILIC chromatography coupled to mass spectrometry reveals important metabolic alterations. J Proteome Res 10: 4825–4834
    1. Gariani K, Menzies KJ, Ryu D, Wegner CJ, Wang X, Ropelle ER, Moullan N, Zhang H, Perino A, Lemos V, Kim B, Park YK, Piersigilli A, Pham TX, Yang Y, Ku CS, Koo SI, Fomitchova A, Canto C, Schoonjans K et al (2016) Eliciting the mitochondrial unfolded protein response by nicotinamide adenine dinucleotide repletion reverses fatty liver disease in mice. Hepatology 63: 1190–1204
    1. Ghaffari P, Mardinoglu A, Nielsen J (2015) Cancer metabolism: a modeling perspective. Front Physiol 6: 382
    1. Hazlehurst JM, Woods C, Marjot T, Cobbold JF, Tomlinson JW (2016) Non‐alcoholic fatty liver disease and diabetes. Metab Clin Exp 65: 1096–1108
    1. Hellerstein MK, Neese RA, Linfoot P, Christiansen M, Turner S, Letscher A (1997) Hepatic gluconeogenic fluxes and glycogen turnover during fasting in humans. A stable isotope study. J Clin Investig 100: 1305–1319
    1. Houtkooper RH, Canto C, Wanders RJ, Auwerx J (2010) The secret life of NAD+: an old metabolite controlling new metabolic signaling pathways. Endocr Rev 31: 194–223
    1. Huang TT, Naeemuddin M, Elchuri S, Yamaguchi M, Kozy HM, Carlson EJ, Epstein CJ (2006) Genetic modifiers of the phenotype of mice deficient in mitochondrial superoxide dismutase. Hum Mol Genet 15: 1187–1194
    1. Hyötyläinen T, Jerby L, Petäjä EM, Mattila I, Jäntti S, Auvinen P, Gastaldelli A, Yki‐Järvinen H, Ruppin E, Orešič M (2016) Genome‐scale study reveals reduced metabolic adaptability in patients with non‐alcoholic fatty liver disease. Nat Commun 7: 8994
    1. Kalhan SC, Guo LN, Edmison J, Dasarathy S, McCullough AJ, Hanson RW, Milburn M (2011) Plasma metabolomic profile in nonalcoholic fatty liver disease. Metabolism 60: 404–413
    1. Karpe F, Dickmann JR, Frayn KN (2011) Fatty acids, obesity, and insulin resistance: time for a reevaluation. Diabetes 60: 2441–2449
    1. Lakshman R, Shah R, Reyes‐Gordillo K, Varatharajalu R (2015) Synergy between NAFLD and AFLD and potential biomarkers. Clin Res Hepatol Gastroenterol 39: S29–S34
    1. Lee S, Mardinoglu A, Lee D, Nielsen J (2016a) Dysregulated signaling hubs of liver lipid metabolism reveal hepatocellular carcinoma pathogenesis. Nucleic Acids Res 44: 5529–5539
    1. Lee S, Zhang C, Kilicarslan M, Bluher M, Uhlen M, Nielsen J, Smith U, Serlie MJ, Boren J, Mardinoglu A (2016b) Integrated network analysis reveals an association between plasma mannose levels and insulin resistance. Cell Metab 24: 172–184
    1. Liao YJ, Chen TL, Lee TS, Wang HA, Wang CK, Liao LY, Liu RS, Huang SF, Chen YM (2012) Glycine N‐methyltransferase deficiency affects Niemann‐Pick type C2 protein stability and regulates hepatic cholesterol homeostasis. Mol Med 18: 412–422
    1. Lofgren L, Stahlman M, Forsberg GB, Saarinen S, Nilsson R, Hansson GI (2012) The BUME method: a novel automated chloroform‐free 96‐well total lipid extraction method for blood plasma. J Lipid Res 53: 1690–1700
    1. Loh K, Deng H, Fukushima A, Cai X, Boivin B, Galic S, Bruce C, Shields BJ, Skiba B, Ooms LM, Stepto N, Wu B, Mitchell CA, Tonks NK, Watt MJ, Febbraio MA, Crack PJ, Andrikopoulos S, Tiganis T (2009) Reactive oxygen species enhance insulin sensitivity. Cell Metab 10: 260–272
    1. Lundbom J, Hakkarainen A, Soderlund S, Westerbacka J, Lundbom N, Taskinen MR (2011) Long‐TE H‐1 MRS suggests that liver fat is more saturated than subcutaneous and visceral fat. NMR Biomed 24: 238–245
    1. Machado MV, Cortez‐Pinto H (2012) Non‐invasive diagnosis of non‐alcoholic fatty liver disease – a critical appraisal. J Hepatol 58: 1007–1019
    1. Mardinoglu A, Nielsen J (2012) Systems medicine and metabolic modelling. J Intern Med 271: 142–154
    1. Mardinoglu A, Agren R, Kampf C, Asplund A, Nookaew I, Jacobson P, Walley AJ, Froguel P, Carlsson LM, Uhlen M, Nielsen J (2013a) Integration of clinical data with a genome‐scale metabolic model of the human adipocyte. Mol Syst Biol 9: 649
    1. Mardinoglu A, Gatto F, Nielsen J (2013b) Genome‐scale modeling of human metabolism – a systems biology approach. Biotechnol J 8: 985–996
    1. Mardinoglu A, Agren R, Kampf C, Asplund A, Uhlen M, Nielsen J (2014a) Genome‐scale metabolic modelling of hepatocytes reveals serine deficiency in patients with non‐alcoholic fatty liver disease. Nat Commun 5: 3083
    1. Mardinoglu A, Kampf C, Asplund A, Fagerberg L, Hallstrom BM, Edlund K, Bluher M, Ponten F, Uhlen M, Nielsen J (2014b) Defining the human adipose tissue proteome to reveal metabolic alterations in obesity. J Proteome Res 13: 5106–5119
    1. Mardinoglu A, Nielsen J (2015) New paradigms for metabolic modeling of human cells. Curr Opin Biotechnol 34: 91–97
    1. Mardinoglu A, Heiker JT, Gartner D, Bjornson E, Schon MR, Flehmig G, Kloting N, Krohn K, Fasshauer M, Stumvoll M, Nielsen J, Bluher M (2015a) Extensive weight loss reveals distinct gene expression changes in human subcutaneous and visceral adipose tissue. Sci Rep 5: 14841
    1. Mardinoglu A, Shoaie S, Bergentall M, Ghaffari P, Zhang C, Larsson E, Bäckhed F, Nielsen J (2015b) The gut microbiota modulates host amino acid and glutathione metabolism in mice. Mol Syst Biol 11: 834
    1. McQuaid SE, Hodson L, Neville MJ, Dennis AL, Cheeseman J, Humphreys SM, Ruge T, Gilbert M, Fielding BA, Frayn KN, Karpe F (2011) Downregulation of adipose tissue fatty acid trafficking in obesity a driver for ectopic fat deposition? Diabetes 60: 47–55
    1. Nestel PJ, Whyte HM (1968) Plasma free fatty acid and triglyceride turnover in obesity. Metab Clin Exp 17: 1122–1128
    1. Norrelund H, Nair KS, Jorgensen JO, Christiansen JS, Moller N (2001) The protein‐retaining effects of growth hormone during fasting involve inhibition of muscle‐protein breakdown. Diabetes 50: 96–104
    1. Patterson BW, Horowitz JF, Wu GY, Watford M, Coppack SW, Klein S (2002) Regional muscle and adipose tissue amino acid metabolism in lean and obese women. Am J Physiol Endocrinol Metab 282: E931–E936
    1. Pozefsky T, Tancredi RG, Moxley RT, Dupre J, Tobin JD (1976) Effects of brief starvation on muscle amino acid metabolism in nonobese man. J Clin Investig 57: 444–449
    1. Ratziu V, Bellentani S, Cortez‐Pinto H, Day C, Marchesini G (2010) A position statement on NAFLD/NASH based on the EASL 2009 special conference. J Hepatol 53: 372–384
    1. Reichard GA Jr, Owen OE, Haff AC, Paul P, Bortz WM (1974) Ketone‐body production and oxidation in fasting obese humans. J Clin Investig 53: 508–515
    1. Senthilkumar R, Sengottuvelan M, Nalini N (2004) Protective effect of glycine supplementation on the levels of lipid peroxidation and antioxidant enzymes in the erythrocyte of rats with alcohol‐induced liver injury. Cell Biochem Funct 22: 123–128
    1. Sim WC, Yin HQ, Choi HS, Choi YJ, Kwak HC, Kim SK, Lee BH (2015) L‐serine supplementation attenuates alcoholic fatty liver by enhancing homocysteine metabolism in mice and rats. J Nutr 145: 260–267
    1. Stahlman M, Fagerberg B, Adiels M, Ekroos K, Chapman JM, Kontush A, Boren J (2013) Dyslipidemia, but not hyperglycemia and insulin resistance, is associated with marked alterations in the HDL lipidome in type 2 diabetic subjects in the DIWA cohort: impact on small HDL particles. Biochim Biophys Acta 1831: 1609–1617
    1. Tamura S, Shimomura I (2005) Contribution of adipose tissue and de novo lipogenesis to nonalcoholic fatty liver disease. J Clin Investig 115: 1139–1142
    1. Uhlen M, Fagerberg L, Hallstrom BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson A, Kampf C, Sjöstedt E, Asplund A, Lundberg E, Djureinovic D, Odeberg J, Habuka M, Tahmasebpoor S, Danielsson A, Edlund K, Szigyarto CA, Skogs M, Takanen JO et al (2015) Tissue‐based map of the human proteome. Science 347: 1260419
    1. Varemo L, Scheele C, Broholm C, Mardinoglu A, Kampf C, Asplund A, Nookaew I, Uhlen M, Pedersen BK, Nielsen J (2015) Proteome‐ and transcriptome‐driven reconstruction of the human myocyte metabolic network and its use for identification of markers for diabetes. Cell Rep 11: 921–933
    1. Vetelainen R, van Vliet A, Gouma DJ, van Gulik TM (2007) Steatosis as a risk factor in liver surgery. Ann Surg 245: 20–30
    1. Wallace JC, Barritt GJ (2005) Gluconeogenesis. Encyclopedia Life Sci doi:
    1. Yizhak K, Gabay O, Cohen H, Ruppin E (2013) Model‐based identification of drug targets that revert disrupted metabolism and its application to ageing. Nat Commun 4: 2632
    1. Yizhak K, Gaude E, Le Devedec S, Waldman YY, Stein GY, van de Water B, Frezza C, Ruppin E (2014a) Phenotype‐based cell‐specific metabolic modeling reveals metabolic liabilities of cancer. Elife 3: e03641
    1. Yizhak K, Le Devedec SE, Rogkoti VM, Baenke F, de Boer VC, Frezza C, Schulze A, van de Water B, Ruppin E (2014b) A computational study of the Warburg effect identifies metabolic targets inhibiting cancer migration. Mol Syst Biol 10: 744
    1. Zhang C, Ji B, Mardinoglu A, Nielsen J, Hua Q (2015) Logical transformation of genome‐scale metabolic models for gene level applications and analysis. Bioinformatics 31: 2324–2331
    1. Zhou CC, Yang X, Hua X, Liu J, Fan MB, Li GQ, Song J, Xu TY, Li ZY, Guan YF, Wang P, Miao CY (2016) Hepatic NAD(+) deficiency as a therapeutic target for non‐alcoholic fatty liver disease in ageing. Br J Pharmacol 173: 2352–2368

Source: PubMed

3
Se inscrever