Elsevier

Alcohol

Volume 46, Issue 8, December 2012, Pages 715-725
Alcohol

Rodent models of alcoholic liver disease: Of mice and men

https://doi.org/10.1016/j.alcohol.2012.08.004Get rights and content

Abstract

Alcoholic liver disease (ALD) is a major cause of acute and chronic liver disease worldwide. The progressive nature of ALD is well described; however, the complex interactions under which these pathologies evolve remain to be fully elucidated. Clinically there are no clear biomarkers or universally accepted, effective treatment strategies for ALD. Experimental models of ALD are an important component in identifying underlying mechanisms of alcohol-induced injury to develop better diagnostic markers, predictors of disease progression, and therapeutic targets to manage, halt, or reverse disease progression. Rodents remain the most accessible model for studying ALD pathology. Effective rodent models must mimic the natural history of ALD while allowing examination of complex interactions between multiple hepatic, and non-hepatic, cell types in the setting of altered metabolic or oxidative/nitrosative stress, inflammatory responses, and sensitivity to cytotoxic stress. Additionally, mode and duration of alcohol delivery influence hepatic response and present unique challenges in understanding disease pathology. This review provides an overview of rodent models of ALD, their strengths and weaknesses relative to human disease states, and provides insight of the potential to develop novel rodent models to simulate the course of human ALD.

Introduction

Alcoholic liver disease (ALD) is a significant cause of morbidity and mortality globally (Beier, Arteel, & McClain, 2011; Gao & Bataller, 2012; Jeong & Gao, 2008; Nath & Szabo, 2009), and refers to a spectrum of hepatic pathologies resulting from acute/binge or chronic alcohol exposure/abuse for which disease progression develops in a dose and time dependent manner (Diehl, 2002; Nath & Szabo, 2009). Alcohol abuse is a leading factor in mortality from liver disease and increases the risk for a wide range of adverse health effects (Beier et al., 2011; Gao & Bataller, 2012; Gerke, Hapke, Rumpf, & John, 1997; Siegmund, Haas, Schneider, & Singer, 2003). In the United States, the Centers for Disease Control and Prevention estimate that ≈50% of people aged 18 or older drink alcohol regularly, and of these, 5% are classified as heavy drinkers (≥30 g alcohol/day) and 15% binge drink (≥5 drinks consumed on a single occasion) (CDC, 2009; Mann, Smart, & Govoni, 2003; Naimi et al., 2003).

The toxic effects of alcohol are exerted on multiple organs; however, the liver, as the primary site of alcohol metabolism, is a major target of injury (Karinch, Martin, & Vary, 2008; Lieber, 2000, 2005). Both chronic and acute drinking deliver unique pathological consequences that affect liver disease and injury, and almost half of all end-stage liver disease in the United States is attributed to alcohol abuse (Beier et al., 2011; Gao & Bataller, 2012). Currently there are no effective, universally accepted therapies for treatment of ALD (Arteel, 2010; Diehl, 2002). Understanding the pathology of ALD is impeded by the complexity of interactions between alcohol (including amount, duration, type consumed) and different hepatic cell types. The effect(s) of alcohol is/are further complicated by host genetics, variability in immunological and metabolic responses, nutritional status, and the presence/absence of comorbid factors such as smoking, and obesity (Nath & Szabo, 2009; Tsukamoto, Machida, Dynnyk, & Mkrtchyan, 2009).

Significant progress has been made in developing animal models with which to investigate mechanisms of ALD initiation and progression. To be effective, ALD models should replicate the etiology and natural history of the human disease. In human ALD pathology the “classic” pattern of liver injury begins with hepatomegaly and alcoholic steatosis (fatty liver), an event that occurs in ≈90% of heavy alcohol users. Of the patients that develop alcohol-induced steatosis, 10–35% progress to steatohepatitis (fatty liver with inflammation), and a further 35–40% go on to develop fibrosis and cirrhosis (end-stage liver disease) (Arteel, 2010; Mann et al., 2003; Nath & Szabo, 2009; Sozio & Crabb, 2008; Szabo & Bala, 2010). Cirrhosis, the most serious form of ALD, is directly associated with duration and amount of alcohol consumed (estimated to be ≈30 g/day for 10 years) (Mann et al., 2003), and is the leading risk factor for subsequent development of hepatocellular carcinoma (HCC) (McKillop & Schrum, 2009).

During early stages of ALD, alcohol consumption diverts metabolic pathways toward hepatic triglyceride accumulation (Lieber, 1991; Sozio & Crabb, 2008). Lipid accumulation leads to increased derangement of metabolic function and increases hepatic sensitivity to toxins (de la Hall et al., 2001; Siegmund et al., 2003). This is an important prerequisite during the development of inflammation (Ramaiah, Rivera, & Arteel, 2004). Metabolic disturbances also contribute to impaired nutrient absorption and distribution, effects that are dependent on both amount of alcohol consumed, and patterns of consumption.

Chronic, long-term alcohol consumption and metabolism is associated with metabolic derangements and changes in nicotinamide adenine dinucleotide (NAD/NADH) ratios favoring accumulation of reducing equivalents in the liver (NADH). Changes in NAD/NADH ratio contribute to hepatic accumulation of triglycerides and depress the citric acid cycle. Alcohol also affects mitochondrial membrane function, metabolic demand, and generation of reactive oxygen species (ROS), a factor exacerbated by alcohol-dependent induction of cytochrome P450 2E1 (CYP2E1) (Lieber, 2005; Lu & Cederbaum, 2008). The toxic responses and injury associated with acute/binge drinking are mediated by amount and rate of alcohol consumption. During acute alcohol consumption, the majority of alcohol is metabolized by successive oxidation reactions, first via alcohol dehydrogenase (ADH) to acetaldehyde, which is in turn oxidized by acetaldehyde dehydrogenase (ALDH) to acetate and water (Lieber, 2005). Acetaldehyde is a toxic intermediate that is usually processed efficiently to prevent accumulation. However, chronic long-term alcohol ab(use) leads to CYP2E1 enzyme induction, which also generates acetaldehyde as the first metabolite during alcohol oxidation (Lieber, 2005; Lu & Cederbaum, 2008). Cytochrome P450 enzymes require oxygen for their catalytic function and reactions mediated by CYP2E1 are poorly coupled, leading to incomplete oxygen reduction and formation of oxyradicals and the partially reduced hydroxyethyl radical, thereby increasing oxidative stress responses (Albano, 2006; Lieber, 2005; Lu & Cederbaum, 2008). In addition, CYP2E1 is associated with increased alcohol tolerance observed in individuals that chronically abuse alcohol and plays a significant role in activation of pro-carcinogens to carcinogens (Dey & Cederbaum, 2006; Lieber, 2005; Lu & Cederbaum, 2008).

In addition to changes in hepatic parenchymal cell (hepatocyte) physiology and function, human ALD is characterized by a state of chronic hepatic inflammation. Alcohol leads to alterations in the gastrointestinal mucosa by disrupting epithelial tight junctions allowing for bacterial endotoxin (lipopolysaccharide; LPS) translocation into the liver via the portal vein (Beck, Morris, & Buell, 1986; Siegmund et al., 2003; Szabo & Bala, 2010). Hepatic response to gut derived endotoxin is a critical step in the development of ALD (Rao, 2009). Presence of LPS in the liver activates innate immune responses primarily via Kupffer cell sensitization (Szabo & Bala, 2010; Thurman, 1998; Yin et al., 1999) leading to intrahepatic inflammation and the production of tumor necrosis factor α (TNFα) and other pro-inflammatory cytokines (Jeong & Gao, 2008; Szabo & Bala, 2010). Additionally innate immune signaling is a mediator of tissue and organ homeostasis regulating proliferation and apoptosis of intestinal epithelial cells, and modulating liver regeneration after loss of liver mass (Seki & Schnabl, 2012). Aberrant regulation of immune system signaling may trigger harmful, inflammatory responses that contribute to tissue and organ injuries, fibrosis, and carcinogenesis (Seki & Schnabl, 2012). Collectively, these cytokine cascades combine to orchestrate liver injury, largely through neutrophil recruitment, although other inflammatory cell types (natural killer, natural killer T-lymphocytes, T-lymphocytes, and dendritic cells) also become activated, each serving unique roles in hepatic injury, repair and remodeling (Gao et al., 2011). Finally, prolonged alcohol-induced changes to liver function, hepatic circulation, and immune responses lead to hepatic stellate cell activation from a quiescent, lipid/vitamin A storing phenotype, to a pro-mitogenic, collagen producing state (de la Hall et al., 2001; Karaa, Thompson, McKillop, Clemens, & Schrum, 2008). Initially, collagen deposition is localized to perivenular and pericellular regions, resulting in portal tract-septal fibrosis that surrounds apoptotic/necrotic hepatocytes (Michalak et al., 2003), and the eventual formation of fibrous septae and scar tissue that encompasses regenerative hepatocytes (Diehl, 2002; Karaa et al., 2008; McKillop, Moran, Jin, & Koniaris, 2006; Ramaiah et al., 2004).

In developing effective models it is important to consider the goal of the intended study, the benefits to scientific research, and the potential limitations with respect to the contribution of the biochemical and pathophysiological consequences of acute or chronic alcohol consumption to organs, tissues and cells, and the role of genetic variability inherent to the population. As is evident from the preceding summary, while multiple factors contribute to ALD progression, there are three essential characteristics related to ALD pathology that must be considered in developing effective disease models. 1. Changes in hepatic metabolism, and particularly changes that lead to accumulation of lipids and/or depletion of essential nutrients, and enhanced hepatotoxicity of alcohol with formation of ROS. 2. Activation of innate inflammatory immune responses, Kupffer cell activation and associated induction of pro-inflammatory cytokines, and migration of infiltrating neutrophils. 3. Increased hepatic damage resulting from persistent inflammatory immune responses leading to stellate cell activation and collagen deposition, combined with compensatory hepatic regeneration.

In addition to these factors increasing evidence suggests other factors may be equally important in considering models of ALD. For example, alcohol induces changes in the gut microbiome that may influence innate and inflammatory immune responses (Seki & Schnabl, 2012; Son, Kremer, & Hines, 2010; Szabo & Bala, 2010; Yan et al., 2011). Indeed gut sterilization by antibiotics or alteration of gut microbiota using probiotics effectively reduces hepatic injury in experimental ALD (Miller & Spicer, 2012; Yan et al., 2011). Additionally, as the liver is a sexually dimorphic organ, the effect(s) of alcohol on liver pathology in males and females is an important factor in hepatic disease pathology (Ellefson et al., 2011; Ramaiah et al., 2004). For instance, while females are more susceptible to initial injury following alcohol consumption they are less likely to progress to cirrhosis and HCC (Frezza et al., 1990; Iimuro et al., 1997; Ikejima et al., 1998; Saunders, Davis, & Williams, 1981; Sorensen et al., 1984).

To date, no rodent model of ALD has been described that effectively replicates human alcoholic hepatitis with progression to fibrosis or cirrhosis without the addition of a secondary insult (e.g. iron, high fat diet, vitamin supplementation, LPS injection) (de la Hall et al., 2001; Karaa et al., 2008; Nanji & French, 2003). The aim of the current review is to provide a summary of the rodent models of ALD currently in use, the relative merits and potential drawbacks associated with these models, and the possible application of newer models being developed that may overcome some, or all, of these limitations.

Section snippets

Animal models of alcohol ingestion

A number of species have been utilized to study ALD, the most common of which are rodents (mice, rats, hamsters), and primates (Batra et al., 1995; Ketcham, Wexler, & Mantel, 1963; Lieber, Leo, Mak, DeCarli, & Sato, 1985; Tsukamoto, Reidelberger, French, & Largman, 1984). Of these model organisms, baboons maintained on alcohol drinking-water for 3–4 years develop ALD through all stages of progression and most closely resemble human ALD pathology (Lieber et al., 1985). However, cost and study

Summary and conclusions

Globally, more than half the world's population consumes alcohol. Attempts to censor this behavior have historically proven unsuccessful; however, the health consequences associated with alcohol consumption remain a significant medical and financial burden. Major advances have been made in our knowledge and understanding of the mechanisms and pathology of ALD. Development of effective animal models is an essential tool in unraveling the interactions and roles of multiple cell types, immune

Financial support

This work was supported, in part by NIH grant AA016858 (IHM).

Disclosures

None of the authors have any conflicts of interest to declare.

References (158)

  • N.J. Grahame et al.

    Blood alcohol concentrations after scheduled access in high-alcohol-preferring mice

    Alcohol

    (2003)
  • S. Gupta et al.

    Transplanted isolated hepatocytes: effect of partial hepatectomy on proliferation of long-term syngeneic implants in rat spleen

    Pathology

    (1987)
  • D.D. Harrison-Findik et al.

    Alcohol metabolism-mediated oxidative stress down-regulates hepcidin transcription and leads to increased duodenal iron transporter expression

    Journal of Biological Chemistry

    (2006)
  • M. Katoh et al.

    Chimeric mice with humanized liver

    Toxicology

    (2008)
  • A. Keegan et al.

    Ethanol-related liver injury in the rat: a model of steatosis, inflammation and pericentral fibrosis

    Journal of Hepatology

    (1995)
  • M.F. Lankford et al.

    Drinking of high concentrations of ethanol versus palatable fluids in alcohol-preferring (P) rats: valid animal model of alcoholism

    Alcohol

    (1991)
  • N. Legrand et al.

    Humanized mice for modeling human infectious disease: challenges, progress, and outlook

    Cell Host & Microbe

    (2009)
  • C.S. Lieber

    Metabolism of alcohol

    Clinics in Liver Disease

    (2005)
  • C.S. Lieber et al.

    Study of agents for the prevention of the fatty liver produced by prolonged alcohol intake

    Gastroenterology

    (1966)
  • C.S. Lieber et al.

    Quantitative relationship between amount of dietary fat and severity of alcoholic fatty liver

    American Journal of Clinical Nutrition

    (1970)
  • S. Lindstedt et al.

    Treatment of hereditary tyrosinaemia type I by inhibition of 4-hydroxyphenylpyruvate dioxygenase

    Lancet

    (1992)
  • W. Liu et al.

    Differential effects of daily-moderate versus weekend-binge alcohol consumption on atherosclerotic plaque development in mice

    Atherosclerosis

    (2011)
  • Y. Lu et al.

    CYP2E1 and oxidative liver injury by alcohol

    Free Radical Biology and Medicine

    (2008)
  • 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 Biology and Medicine

    (2010)
  • R.S. McCuskey et al.

    Ethanol binging exacerbates sinusoidal endothelial and parenchymal injury elicited by acetaminophen

    Journal of Hepatology

    (2005)
  • I.H. McKillop et al.

    Molecular pathogenesis of hepatocellular carcinoma

    Journal of Surgical Research

    (2006)
  • I.H. McKillop et al.

    Altered expression of mitogen-activated protein kinases in a rat model of experimental hepatocellular carcinoma

    Hepatology

    (1997)
  • Y. Adachi et al.

    Inactivation of Kupffer cells prevents early alcohol-induced liver injury

    Hepatology

    (1994)
  • E. Albano

    Alcohol, oxidative stress and free radical damage

    Proceedings of the Nutrition Society

    (2006)
  • B.N. Ames et al.

    Oxidants, antioxidants, and the degenerative diseases of aging

    Proceedings of the National Academy of Sciences of the United States of America

    (1993)
  • A.R. Aroor et al.

    Elevated activation of ERK1 and ERK2 accompany enhanced liver injury following alcohol binge in chronically ethanol-fed rats

    Alcoholism: Clinical & Experimental Research

    (2011)
  • G. Arteel

    Animal models of alcoholic liver disease

    Digestive Diseases

    (2010)
  • S.C. Batra et al.

    Gastric metabolism of ethanol in Syrian golden hamster

    Digestive Diseases and Sciences

    (1995)
  • J.I. Beier et al.

    Advances in alcoholic liver disease

    Current Gastroenterology Reports

    (2011)
  • C.H. Best et al.

    Liver damage produced by feeding alcohol or sugar and its prevention by choline

    British Medical Journal

    (1949)
  • E. Brandish et al.

    Drinking patterns and the risk of serious liver disease

    Expert Review of Gastroenterology and Hepatology

    (2010)
  • E. Brandon-Warner et al.

    Chronic ethanol feeding accelerates hepatocellular carcinoma progression in a sex-dependent manner in a mouse model of hepatocarcinogenesis

    Alcoholism: Clinical & Experimental Research

    (2012)
  • K.M. Braun et al.

    Liver disease and compensatory growth: unexpected lessons from genetically altered mice

    International Journal of Developmental Biology

    (1998)
  • J. Brick et al.

    Intoxication is not always visible: an unrecognized prevention challenge

    Alcoholism: Clinical & Experimental Research

    (2009)
  • L.G. Carr et al.

    A quantitative trait locus for alcohol consumption in selectively bred rat lines

    Alcoholism: Clinical & Experimental Research

    (1998)
  • CDC

    Behavioral risk factor surveillance system survey data

    (2009)
  • C. Chen et al.

    Alcohol consumption among young adults ages 18–24 in the United States: results from the 2001–2002 NESARC survey

    Alcohol Research and Health

    (2004–2005)
  • A.A. Chen et al.

    Humanized mice with ectopic artificial liver tissues

    Proceedings of the National Academy of Sciences of the United States of America

    (2011)
  • J.A. Chester et al.

    Effects of stress on alcohol consumption in rats selectively bred for high or low alcohol drinking

    Alcoholism: Clinical & Experimental Research

    (2004)
  • C. Cheung et al.

    Humanized mouse lines and their application for prediction of human drug metabolism and toxicological risk assessment

    Journal of Pharmacology and Experimental Therapeutics

    (2008)
  • S. Copeland et al.

    Acute inflammatory response to endotoxin in mice and humans

    Clinical and Diagnostic Laboratory Immunology

    (2005)
  • J.C. Crabbe et al.

    Preclinical studies of alcohol binge drinking

    Annals of the New York Academy of Sciences

    (2011)
  • S.M. DeNucci et al.

    Rat strain differences in susceptibility to alcohol-induced chronic liver injury and hepatic insulin resistance

    Gastroenterology Research and Practice

    (2010)
  • A. Dey et al.

    Alcohol and oxidative liver injury

    Hepatology

    (2006)
  • M. Dorner et al.

    A genetically humanized mouse model for hepatitis C virus infection

    Nature

    (2011)
  • Cited by (113)

    • Fatty Liver Disease

      2023, MacSween's Pathology of the Liver, Eighth Edition
    View all citing articles on Scopus
    View full text