Elsevier

Free Radical Biology and Medicine

Volume 143, 1 November 2019, Pages 127-139
Free Radical Biology and Medicine

Original article
Glutathione deficiency-elicited reprogramming of hepatic metabolism protects against alcohol-induced steatosis

https://doi.org/10.1016/j.freeradbiomed.2019.07.025Get rights and content

Highlights

  • GSH-deficient (Gclm-KO) liver is acetyl-CoA and glutamate enriched.

  • Low GSH promotes pyruvate and fatty acid oxidation and inhibits lipogenesis.

  • Ethanol-exposed WT and KO liver show differential changes in acetyl-CoA metabolism.

  • Transcriptional and post-transcriptional mechanisms underlie metabolic adaptions.

Abstract

Depletion of glutathione (GSH) is considered a critical pathogenic event promoting alcohol-induced lipotoxicity. We recently show that systemic GSH deficiency in mice harboring a global disruption of the glutamate-cysteine ligase modifier subunit (Gclm) gene confers protection against alcohol-induced steatosis. While several molecular pathways have been linked to the observed hepatic protection, including nuclear factor erythroid 2-related factor 2 and AMP-activated protein kinase pathways, the precise mechanisms are yet to be defined. In this study, to gain insights into the molecular mechanisms underpinning the protective effects of loss of GCLM, global profiling of hepatic polar metabolites combined with liver microarray analysis was carried out. These inter-omics analyses revealed both low GSH- and alcohol-driven changes in multiple cellular pathways involving the metabolism of amino acids, fatty acid, glucose and nucleic acids. Notably, several metabolic changes were uniquely present in alcohol-treated Gclm-null mouse livers, including acetyl-CoA enrichment and diversion of acetyl-CoA flux from lipogenesis to alterative metabolic pathways, elevation in glutamate concentration, and induction of the glucuronate pathway and nucleotide biosynthesis. These metabolic features reflect low GSH-elicited cellular response to chronic alcohol exposure, which is beneficial for the maintenance of hepatic redox and metabolic homeostasis. The current study indicates that fine-tuning of hepatic GSH pool may evoke metabolic reprogramming to cope with alcohol-induced cellular stress.

Introduction

Excessive consumption of alcohol is a major cause of chronic liver disease. Globally, alcoholic liver disease (ALD) accounts for 0.9% of total mortality and 0.6% of disability-adjusted life years and remains a public health problem worldwide [1]. ALD initially manifests as simple fatty liver (steatosis), which may progress to steatohepatitis, liver fibrosis and cirrhosis, and eventually hepatocellular carcinoma. To date, the management of ALD remains challenging due to the lack of detailed understanding of determinants of its pathogenesis and progression.

Chronic alcohol consumption modulates numerous cellular pathways in the liver and other organs, among which alcohol-induced redox perturbation appears to play a critical role in the pathogenesis of ALD [2]. Over-production of reactive molecules, including electrophiles (e.g. acetaldehyde and lipid peroxidation byproducts), reactive oxygen species (ROS) and reactive nitrogen species (RNS), may arise from ethanol metabolism, CYP2E1 induction, mitochondrial dysfunction, and proinflammatory processes [2]. In addition, ethanol exposure was shown to reduce the antioxidant capacity of the liver through depleting antioxidants and inactivating antioxidant enzymes [2]. The pathophysiological consequences of oxidative insults include, among others, impaired lipid metabolism leading to steatosis, hepatocyte injury and activation of liver fibrosis, all of which are features of ALD [3].

It is well established that glutathione (GSH), the most abundant cellular non-protein thiol, plays a pivotal role in maintaining redox homeostasis and contributes significantly to protection against oxidative insults [4]. The rate-limiting step in the GSH biosynthesis pathway is catalyzed by the glutamate-cysteine ligase (GCL), a heterodimer comprising a catalytic (GCLC) and a modifier (GCLM) subunit [5]. Disruption of the mouse Gclc gene in hepatocytes results in >95% depletion of hepatic GSH and induces liver pathologies characteristic of various clinical stages of fatty liver disease [6,7]. Global disruption of mouse Gclm gene generates a mouse model (Gclm-null) that exhibits normal liver functioning in spite of the hepatic GSH concentration being only ~15% of normal [8]. Intriguingly, following 6 week of ethanol intake with the Lieber-DeCarli (LD) diet, Gclm-null mice were protected from alcohol-induced steatosis [9]. Such protection appeared to be associated with redox activation of nuclear factor erythroid 2-related factor 2 (NRF2) and AMP-activated protein kinase (AMPK), which are key regulators of cellular stress response, cellular metabolism and biogenesis [9]. Although these results revealed the hepatoprotective effect of low GSH, the mechanistic details remain largely unknown.

It was documented in ALD patients and experimental animals that alcohol consumption impacts multiple biochemical pathways [10]. In addition to aberrant lipid metabolism, alcohol-related dysregulation of other cellular metabolisms contributes to ALD pathogenesis [10]. In the current study, we aimed to gain insights into the mechanism(s) by which GSH deficiency (due to loss of GCLM) regulates hepatic metabolic homeostasis in response to chronic ethanol consumption. This was accomplished through global profiling of hepatic polar metabolome. Liver microarray analysis was conducted in parallel to explore molecular mechanisms underlying protection from steatosis through integrated pathway analysis.

Section snippets

Reagents

All chemicals and reagents were purchased from Sigma-Aldrich (St. Louis, MO, USA) unless otherwise specified.

Animals and chronic ethanol feeding

All animal experiments were performed at the University of Colorado Anschutz Medical Campus, as reported previously [9]. All animal procedures were approved by and conducted in compliance with Institutional Animal Care and Use Committee (IACUC) of the same institution. Briefly, 10–12 week male C57BL/6J wild-type (WT) and Gclm-null (KO) mice were fed a modified LD diet (Bio-Serv,

Liver metabolic profiling reveals a distinct metabolic signature associated with loss of GCLM and ethanol consumption

During the 6-week feeding period, the daily intake of CON or EtOH diet by WT and Gclm-KO mice were no different (data not shown). The overall liver polar metabolic signature was analyzed by principal components analysis. The scores-scatter plot for liver weight-normalized metabolic signature recorded in positive (Fig. 1A, left panel) and negative (Fig. 1A, right panel) ESI modes showed that mice segregate along first principal component according to their genotypes, irrespective of EtOH

Discussion

There is a large body of evidence indicating that low hepatic GSH content is pathogenically involved in liver diseases of various etiologies [[27], [28], [29], [30]]. This is expected given the crucial function of GSH in maintaining cellular redox homeostasis and in detoxifying electrophiles. New knowledge derived from redox biology research, however, provides convincing evidence that chronic oxidative and nitrosative stress under physiological conditions trigger cellular mechanisms that

Conclusion

The present study has identified new hepatic metabolic signatures intrinsically linked to the loss of GCLM (and by extension low hepatic GSH) and modulated by alcohol consumption, the combination of which are, at least in part, causally associated with hepatic protection against alcohol-induced steatosis in Gclm-KO mice (Fig. 7). These metabolic changes cover multiple biochemical pathways and represent a metabolic network that appears to be reprogramed by both transcriptional and

Conflicts of interest

None.

Funding sources

This work was supported in part by the National Institutes of Health grants K01AA025093 (YC), R24AA022057 (VV), U01AA021724 (VV), the NIH Intramural Research Program (FJG) and the Department of Atomic Energy, Government of India (SKM).

Acknowledgement

We thank Dr. Peng Gang (Yale School of Public Health) for valuable input into our statistical analysis.

References (54)

  • A. Rowland et al.

    The UDP-glucuronosyltransferases: their role in drug metabolism and detoxification

    Int. J. Biochem. Cell Biol.

    (2013)
  • Y. Lu et al.

    Chronic alcohol-induced liver injury and oxidant stress are decreased in cytochrome P4502E1 knockout mice and restored in humanized cytochrome P4502E1 knock-in mice

    Free Radical Biol. Med.

    (2010)
  • Y. Chen et al.

    Glutathione defense mechanism in liver injury: insights from animal models

    Food Chem. Toxicol.: Int. J. Publ. Brit. Ind. Biol. Res. Assoc.

    (2013)
  • B.U. Bradford et al.

    Metabolomic profiling of a modified alcohol liquid diet model for liver injury in the mouse uncovers new markers of disease

    Toxicol. Appl. Pharmacol.

    (2008)
  • X. Shi et al.

    Identification of N-acetyltaurine as a novel metabolite of ethanol through metabolomics-guided biochemical analysis

    J. Biol. Chem.

    (2012)
  • A.I. Cederbaum

    Alcohol metabolism

    Clin. Liver Dis.

    (2012)
  • H. Kaeferstein

    Forensic relevance of glucuronidation in phase-II-metabolism of alcohols and drugs

    Leg. Med.

    (2009)
  • H.G. Gika et al.

    Investigation of chronic alcohol consumption in rodents via ultra-high-performance liquid chromatography-mass spectrometry based metabolite profiling

    J. Chromatogr. A

    (2012)
  • Y. Chen et al.

    Hepatocyte-specific Gclc deletion leads to rapid onset of steatosis with mitochondrial injury and liver failure

    Hepatology

    (2007)
  • Y. Chen et al.

    Chronic glutathione depletion confers protection against alcohol-induced steatosis: implication for redox activation of AMP-activated protein kinase pathway

    Sci. Rep.

    (2016)
  • G.G. Harrigan et al.

    Metabolomics in alcohol research and drug development

    Alcohol Res. Health

    (2008)
  • E.G. Bligh et al.

    A rapid method of total lipid extraction and purification

    Can. J. Biochem. Physiol.

    (1959)
  • Y. Benjamini et al.

    Controlling the False Discovery rate - a practical and powerful approach to multiple testing

    J. Roy. Stat. Soc. B Met.

    (1995)
  • P. Shannon et al.

    Cytoscape: a software environment for integrated models of biomolecular interaction networks

    Genome Res.

    (2003)
  • Y.S. Jung

    Metabolism of sulfur-containing amino acids in the liver: a link between hepatic injury and recovery

    Biol. Pharm. Bull.

    (2015)
  • A. Meister et al.

    New aspects of glutathione metabolism and translocation in mammals

    Ciba Found. Symp.

    (1979)
  • K.K. Kharbanda

    Role of transmethylation reactions in alcoholic liver disease

    World J. Gastroenterol.

    (2007)
  • Cited by (17)

    • Thymol ameliorates ethanol-induced hepatotoxicity via regulating metabolism and autophagy

      2023, Chemico-Biological Interactions
      Citation Excerpt :

      These results indicated that treatment with 100 mM ethanol, 100 μM thymol or 100 mM ethanol plus 100 μM thymol does not induce significant toxic effects on AML12 cells. Ethanol oxidizing system promotes free radical generation [14,19]. We determined the content of ROS using DCFH-DA.

    • Persistence of improved glucose homeostasis in Gclm null mice with age and cadmium treatment

      2022, Redox Biology
      Citation Excerpt :

      This finding stands in contrast to mice with hepatocyte-specific Txnrd1-knockdown (another model of chronic thiol insufficiency), which display increased hepatic glycogen storage [30]. Derivation methods notwithstanding, all current Gclm null mouse models display increased activity of the oxidative stress-responsive and metabolic reprogramming transcription factor Nrf2 and subsequently, increased expression of Nrf2-regulated genes [18,20,27,31]. The lean phenotype observed in Gclm null mice likely results from a switch in hepatic energy utilization in response to GSH deficiency.

    • Integrated non-targeted lipidomics and metabolomics analyses for fluctuations of neonicotinoids imidacloprid and acetamiprid on Neuro-2a cells

      2021, Environmental Pollution
      Citation Excerpt :

      A close relationship possibly exists between the metabolisms of GSH and lipid in living organisms. A previous study reported the depletion of GSH as a critical pathogenic event that can promote alcohol-induced lipotoxicity (Chen et al., 2019). Glycerophospholipid metabolism, GSH metabolism, and nucleotide metabolism pathways can be potential biomarkers of the degree of response of S. intermedius larvae to the negative effects of seawater acidification (Li et al., 2020a).

    View all citing articles on Scopus
    1

    Contributed equally.

    2

    Current address: Yale Center for Genome Analysis, Yale School of Medicine, Orange, CT 06477, USA.

    View full text