Elsevier

Alcohol

Volume 91, March 2021, Pages 1-9
Alcohol

Dihydromyricetin improves mitochondrial outcomes in the liver of alcohol-fed mice via the AMPK/Sirt-1/PGC-1α signaling axis

https://doi.org/10.1016/j.alcohol.2020.10.002Get rights and content

Highlights

  • Dihydromyricetin (DHM), a bioactive flavonoid, improved mitochondrial outcomes in the liver of male C57BL/6J mice after chronic alcohol feeding.

  • Alcohol inhibition of key metabolic enzymes, AMPK and Sirtuins, and mitochondrial injury are reversed with DHM administration.

  • These findings support the utility of DHM, a dietary supplement, as a novel therapeutic for the reduction/prevention of alcohol-related mitochondrial injury in the liver.

  • This dietary flavonoid provides key metabolic responses that support its use for other mitochondria-related disorders.

Abstract

Alcoholic liver disease (ALD), due to the multifactorial damage associated with alcohol (ethanol) consumption and metabolism, is one of the most prevalent liver diseases in the United States. The liver is the primary site of ethanol metabolism and is subsequently injured due to the production of reactive oxygen species (ROS), acetaldehyde, and metabolic stress. Building evidence suggests that dihydromyricetin (DHM), a bioactive flavonoid isolated from Hovenia dulcis, provides hepatoprotection by enhancing ethanol metabolism in the liver by maintaining hepatocellular bioenergetics, reductions of oxidative stress, and activating lipid oxidation pathways. The present study investigates the utility of DHM on hepatic mitochondrial biogenesis via activation of the AMP-activated protein kinase (AMPK)/Sirtuin (Sirt)-1/PPARG coactivator 1 (PGC)-1α signaling pathway. We utilized a forced drinking ad libitum study that chronically fed 30% ethanol to male C57BL/6J mice over 8 weeks and induced ALD pathology. We found that chronic ethanol feeding resulted in the suppression of AMPK activation and cytoplasmic Sirt-1 and mitochondrial Sirt-3 expression, effects that were reversed with daily DHM administration (5 mg/kg; intraperitoneally [i.p.]). Chronic ethanol feeding also resulted in hepatic hyperacetylation of PGC-1α, which was improved with DHM administration and its mediated increase of Sirt-1 activity. Furthermore, ethanol-fed mice were found to have increased expression of mitochondrial transcription factor A (TFAM), reduced mitochondrial content as assessed by mitochondrial DNA to nuclear DNA ratios, and significantly lower levels of hepatic ATP. In contrast, DHM administration significantly increased TFAM expression, hepatic ATP concentrations, and induced mitochondrial expression of respiratory complex III and V. In total, this work demonstrates a novel mechanism of DHM that improves hepatic bioenergetics, metabolic signaling, and mitochondrial viability, thus adding to the evidence supporting the use of DHM for treatment of ALD and other metabolic disorders.

Introduction

Alcoholic liver disease (ALD) is one of the most prevalent liver diseases in the United States, and it includes a spectrum of diseases ranging from reversible fatty liver to alcoholic hepatitis, and cirrhosis (Mellinger et al., 2018). The majority of patients diagnosed with ALD are also suffering from alcohol use disorder (AUD), where this latter condition affects over 17 million people in the United States. A concerning trend is the increased incidences of younger adults being diagnosed with ALD due to increasing rates of alcohol (ethanol) abuse. The mechanisms involved in the development of ALD are multifaceted, and it is becoming evident that the disease spectrum and subsequent progress to late-stage ALD with prolonged ethanol abuse results from the multifactorial injurious responses that occur throughout the body (Rehm et al., 2010; Rehm, Samokhvalov, & Shield, 2013; Seitz et al., 2018). Ongoing cycles of high levels of ethanol abuse and ethanol metabolism represent a primary mechanism of organ damage and is one major contributing factor resulting in the pathology of ALD and subsequent development of late-stage ALD (Han et al., 2012).

Ethanol is primarily metabolized in the liver by cytosolic alcohol dehydrogenase (ADH) and the inducible cytochrome P450 2E1 (CYP2E1) in mitochondria and endoplasmic reticulum (Lieber, Rubin, & DeCarli, 1970; Seitz et al., 2018). The metabolism of ethanol by ADH1 and CYP2E1 results in the oxidation of ethanol to acetaldehyde (ACH), a highly reactive and toxic metabolite, which is then further metabolized to acetate by aldehyde dehydrogenase (ALDH2) located in the mitochondria in the liver. Throughout this metabolic process, reactive oxygen species (ROS) formation occurs, with CYP2E1 oxidation contributing to much of the ROS stress in the liver (Leung & Nieto, 2013; Lieber, 1997; Neve & Ingelman-Sundberg, 2000). The combination of high rates of ethanol metabolism, increased ROS, and production of ACH results in multiple responses in the liver that dysregulate energy signaling and lipid metabolic pathways, alongside the induction of inflammatory responses (Ceni, Mello, & Galli, 2014; Seitz et al., 2018). Additionally, ACH and ROS alter mitochondrial structure and activity, thereby leading to functional impairment, including decreased oxidative phosphorylation (ATP generation), ROS exacerbation, and a decrease in ALDH2 activity, resulting in elevated ACH (O'Shea et al., 2010). Furthermore, the nicotinamide adenine dinucleotide (NAD)-dependent metabolism of ethanol/ACH, which results in the depletion of hepatic NAD+, contributes to additional metabolic stressors (French, 2016; Wang et al., 2018). In an effort to adapt to the ongoing hepatic injury from high levels of ethanol exposure, mitochondria undergo various responses, such as biogenesis, to maintain mitochondrial integrity and prevent further damage resulting from dysfunctional metabolic responses (Degli Esposti et al., 2012; Pessayre et al., 2012; Serviddio, Bellanti, Sastre, Vendemiale, & Altomare, 2010). Collectively, this work suggests that pharmacological agents that target these aspects of ethanol-mediated stress on mitochondria could be critical in protecting hepatocytes from energy impairments that result from mitochondrial-related stress in the liver.

One key regulator of mitochondrial biogenesis and cellular energy metabolism is the peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (PGC-1α), which belongs to the family of PGC-1 transcription coactivators (Handschin, 2009; Scarpulla, 2011). PGC-1α is present at low but inducible levels in the liver, where it also regulates most metabolic pathways, such as fatty acid β-oxidation, gluconeogenesis, and ketogenesis (Puigserver et al., 2003; Rhee et al., 2003; Yoon et al., 2001). Under normal physiological stress conditions (e.g., energy deprivation, fasting, and/or low temperatures), PGC-1α is activated via cyclic AMP response element-binding protein (CREB) and post-translationally via adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and NAD+-dependent sirtuin (SIRT)-1 deacetylation (Scarpulla, 2011). Notably, ethanol metabolism inhibits the AMPK-dependent phosphorylation of PGC-1α necessary for activation, and depletes NAD+ concentrations critical for Sirt-1-driven deacetylation, thereby inhibiting PGC-1α activation via ROS stress and energy depletion (Chaung, Jacob, Ji, & Wang, 2008; French, 2016; You, Jogasuria, Taylor, & Wu, 2015). This inhibition of mitochondrial biogenesis, in addition to mitochondrial stress and damage, results in the accumulation of dysfunctional mitochondria and reduced hepatic ability to selectively remove mitochondria by mitophagy (Eid, Ito, Maemura, & Otsuki, 2013; Williams, Ni, Ding, & Ding, 2015).

Dihydromyricetin (DHM), an active bioflavonoid isolated from Hovenia dulcis, has been used in Chinese traditional medicines for centuries, and has been shown to protect the liver against chemically induced liver damage via increased Sirt-1 signaling (Ma et al., 2019). In support of this hypothesis, we and others have shown that DHM can have multiple medicinal benefits, including anti-inflammatory, antioxidant, hepatoprotective, and anti-alcohol properties (Chen et al., 2015; Martínez-Coria, Mendoza-Rojas, Arrieta-Cruz, & López-Valdés, 2019; Qi et al., 2012; Shen et al., 2012; Silva et al., 2020). As part of this later effort, we found that DHM enhances ethanol metabolism in the liver, in part due to increased NAD+ concentrations. Moreover, we found that DHM can reduce hepatic lipid accumulation via AMPK activation in ethanol-fed male C57BL/6J mice (Silva et al., 2020). However, much remains to be elucidated regarding the mechanism by which these metabolic changes in response to ethanol occur. The current study addresses this issue by investigating the hepatoprotective role of DHM in the AMPK/Sirt-1/PGC-1α signaling axis and subsequent mitochondrial viability that is otherwise altered by chronic ethanol feeding.

Section snippets

Animals and experimental design

A forced drinking ad libitum study was conducted as previously described using 8-week-old male C57BL/6J mice and the provision of a single bottle of ethanol [ethanol-fed], starting at 5% and gradually increasing to 30% ethanol, or tap water [water-fed controls], for a total of 8 weeks (Brandon-Warner, Schrum, Schmidt, & McKillop, 2012; Keegan, Martini, & Batey, 1995; Silva et al., 2020). Mice were grouped as follows: 1) water-fed + saline intraperitoneal (i.p.) injections (n = 6), 2)

DHM reverses ethanol-mediated inhibition of hepatic AMPK and increases Sirt-1 and mitochondrial Sirt-3 expression

We evaluated the expression of cytoplasmic Sirt-1, activated pAMPK (Thr172) relative to total AMPK, mitochondrial Sirt-3, and voltage-dependent anion channel 1 (VDAC1) in the liver of chronic ethanol-fed and water-fed mice. Ethanol feeding significantly reduced Sirt-1 and Sirt-3 expression by 37% and 31%, respectively (Fig. 1). Additionally, chronic ethanol feeding inhibited the activation of AMPK (phosphorylation at Thr172) by 43% compared to water-fed controls (†p < 0.05 vs. water-fed; Fig. 1

Discussion

The outcomes from the present work represent the first demonstration that DHM pharmacologically preserves mitochondrial viability following ethanol feeding that suppresses biogenesis and induced injury. These data support the hypothesis that DHM pharmacological effects on NAD bioenergetics result in the activation of the AMPK/Sirt-1/PGC-1α axis and subsequent modification of the expression of mitochondrial respiratory complex proteins III and V and ATP production. Beyond the hepatoprotective

Author statement

Joshua Silva: Original draft preparation, methodology, data analysis, figure design. Maximilian H. Spatz: Methodology, data analysis, and review. Carson Folk: Methodology, data analysis, and review. Arnold Chang: Methodology, data analysis, and review. Enrique Cadenas: Scientific conceptualization, original draft preparation, editing, and data interpretation. Jing Liang: Original draft preparation, editing, and data interpretation. Daryl Davies: Scientific conceptualization, writing – original

Acknowledgments

This work was supported by funding and grants from the NIH NIAA R01AA022448, USC GoodNeighbors Campaign, USC School of Pharmacy, and American Foundation for Pharmaceutical Education (AFPE).

References (61)

  • C.S. Lieber

    Metabolism of alcohol

    Clinics in Liver Disease

    (2005)
  • C.S. Lieber et al.

    Hepatic microsomal ethanol oxidizing system (MEOS): Differentiation from alcohol dehydrogenase and NADPH oxidase

    Biochemical and Biophysical Research Communications

    (1970)
  • J.Q. Ma et al.

    Ampelopsin attenuates carbon tetrachloride-induced mouse liver fibrosis and hepatic stellate cell activation associated with the SIRT1/TGF-β1/Smad3 and autophagy pathway

    International Immunopharmacology

    (2019)
  • E.P. Neve et al.

    Molecular basis for the transport of cytochrome P450 2E1 to the plasma membrane

    Journal of Biological Chemistry

    (2000)
  • A. Picca et al.

    Regulation of mitochondrial biogenesis through TFAM-mitochondrial DNA interactions. Useful insights from aging and calorie restriction studies

    Mitochondrion

    (2015)
  • S. Qi et al.

    Ampelopsin reduces endotoxic inflammation via repressing ROS-mediated activation of PI3K/Akt/NF-κB signaling pathways

    International Immunopharmacology

    (2012)
  • J. Rehm et al.

    Global burden of alcoholic liver diseases

    Journal of Hepatology

    (2013)
  • R.C. Scarpulla

    Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network

    Biochimica et Biophysica Acta

    (2011)
  • S. Wang et al.

    Nicotinamide riboside attenuates alcohol induced liver injuries via activation of SirT1/PGC-1α/mitochondrial biosynthesis pathway

    Redox Biology

    (2018)
  • H. Yin et al.

    Deletion of SIRT1 from hepatocytes in mice disrupts lipin-1 signaling and aggravates alcoholic fatty liver

    Gastroenterology

    (2014)
  • H. Yin et al.

    MicroRNA-217 promotes ethanol-induced fat accumulation in hepatocytes by down-regulating SIRT1

    Journal of Biological Chemistry

    (2012)
  • K. Aquilano et al.

    P53 orchestrates the PGC-1α-mediated antioxidant response upon mild redox and metabolic imbalance

    Antioxidants and Redox Signaling

    (2013)
  • V. Audrito et al.

    NAMPT and NAPRT: Two metabolic enzymes with key roles in inflammation

    Frontiers in Oncology

    (2020)
  • C. Cantó et al.

    AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity

    Nature

    (2009)
  • E. Ceni et al.

    Pathogenesis of alcoholic liver disease: Role of oxidative metabolism

    World Journal of Gastroenterology

    (2014)
  • W.W. Chaung et al.

    Suppression of PGC-1alpha by ethanol: Implications of its role in alcohol induced liver injury

    International Journal of Clinical and Experimental Medicine

    (2008)
  • S.E. Choi et al.

    Elevated microRNA-34a in obesity reduces NAD+ levels and SIRT1 activity by directly targeting NAMPT

    Aging Cell

    (2013)
  • D. Degli Esposti et al.

    Mitochondrial roles and cytoprotection in chronic liver injury

    Biochemistry Research International

    (2012)
  • R.B. Ding et al.

    Emerging roles of SIRT1 in fatty liver diseases

    International Journal of Biological Sciences

    (2017)
  • N. Eid et al.

    Elevated autophagic sequestration of mitochondria and lipid droplets in steatotic hepatocytes of chronic ethanol-treated rats: An immunohistochemical and electron microscopic study

    Journal of Molecular Histology

    (2013)
  • Cited by (32)

    • Dietary modulation and mitochondrial DNA damage

      2023, Molecular Nutrition and Mitochondria: Metabolic Deficits, Whole-Diet Interventions, and Targeted Nutraceuticals
    • Huang Bai Jian Pi decoction alleviates diarrhea and represses inflammatory injury via PI3K/Akt/NF-κB pathway: In vivo and in vitro studies

      2022, Journal of Ethnopharmacology
      Citation Excerpt :

      In diarrheal cases in animals, the dysfunction of the large intestine is destroyed usually, and the lungs share an interior-exterior relationship with the large intestine in TCM, which could be one of causes in lung lesions (Hua et al., 2020). Liver disorder could have resulted from the high-sugar-high-fat diet and alcohol (Silva et al., 2021). The HBJP decoction alleviated organ injuries of rats in the model group.

    • Molecular mechanisms and therapeutic implications of dihydromyricetin in liver disease

      2021, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      The proposed mechanism was that DHM activates the antioxidant systems by increasing the expression of Nrf2 as well as its antioxidant product heme oxygenase-1 (HO-1), while suppressing intracellular ROS generation and the hepatic expression of reactive oxygen species (ROS)-producing CYP2E1 enzyme [31]. Similarly, Silva et al. [32] reported that DHM administration reversed the suppression of adenosine monophosphate-activated protein kinase (AMPK) activation and Sirtuins (Sirt1 and Sirt3) expression in chronic ALD models, and improved hepatic hyperacetylation of peroxisome proliferator activated receptor γ (PPARγ) coactivator 1α (PGC-1α) and the expression of mitochondrial transcription factor A (TFAM), which resulted in the enhancement of mitochondrial biogenesis. In addition, Qiu et al. [33] further revealed the crosstalk between p62-mediated autophagy and Nrf2 in the alleviation of alcoholic liver disease symptoms by DHM.

    View all citing articles on Scopus
    View full text