Sie sind auf Seite 1von 11

REVIEWS

Alcoholic liver disease: pathogenesis and new


targets for therapy
José Altamirano and Ramón Bataller
Abstract | Alcoholic liver disease (ALD) is a major cause of morbidity and mortality worldwide. The spectrum
of disease ranges from fatty liver to hepatic inflammation, necrosis, progressive fibrosis and hepatocellular
carcinoma. In developed countries, ALD is a major cause of end-stage liver disease that requires
transplantation. The most effective therapy for ALD is alcohol abstinence. However, for patients with severe
forms of ALD (that is, alcoholic hepatitis) and for those who do not achieve abstinence from alcohol, targeted
therapies are urgently needed. The development of new drugs for ALD is hampered by the scarcity of studies
and the drawbacks of existing animal models, which do not reflect all the features of the human disease.
However, translational research using liver samples from patients with ALD has identified new potential
therapeutic targets, such as CXC chemokines, osteopontin and tumor necrosis factor receptor superfamily
member 12A. The pathogenetic roles of these targets, however, remain to be confirmed in animal models. This
Review summarizes the epidemiology, natural history, risk factors and current knowledge of the pathogenetic
mechanisms of ALD. In addition, this article provides a detailed description of the findings of these
translational studies and of the animal models used to study ALD.
Altamirano, J. & Bataller, R. Nat. Rev. Gastroenterol. Hepatol. 8, 491–501 (2011); published online 9 August 2011; doi:10.1038/nrgastro.2011.134

Introduction
Sustained, excessive alcohol consumption is an addictive drug companies and the drawbacks of current experi­
behavior disorder that occurs in all ethnicities and age mental models of ALD. Moreover, public funding for
groups, and in both sexes.1,2 In many individuals, consis­ research into alcohol-related disorders has been scarce,
tently high levels of alcohol intake leads to alcoholic liver especially in the field of hepatology.
disease (ALD), a major cause of morbidity and mortal­ In this Review, we describe the epidemiology and
ity worldwide. Heavy alcohol drinking is also associated natural history of ALD, as well as the risk factors associ­
with disorders unrelated to the liver, such as infections, ated with this disease. In addition, we present the current
malignancies, cardiovascular events and diseases of the knowledge of the pathogenetic mechanisms underlying
nervous system, pancreas and kidneys.3 ALD. This article also includes detailed descriptions
The spectrum of ALD encompasses fatty liver, hepatic of translational studies conducted using human tissue
inflammation and necrosis, progressive fibrosis and samples, which have identified potential new therapeutic
hepatocellular carcinoma.2 Furthermore, a sustained, targets for ALD, and the animal models currently used
excessive alcohol intake favors the progression of other to study this disease.
liver diseases, such as chronic viral hepatitis (hepa­
titis C and B) and other metabolic liver diseases, such as Epidemiology of ALD
hemochromatosis, Wilson disease and fatty liver associ­ Prevalence
ated with the metabolic syndrome.4–8 Despite the eco­ Alcohol abuse is a major cause of preventable morbid­
Liver Unit, Hospital
nomic burden of ALD and its severe effect on health, ity and mortality worldwide. According to the WHO, Clínic, Institut
few advances have been made in the management of morbidity from alcohol misuse in developed countries d’Investigacions
Biomèdiques August Pi
patients with this condition.2 No modern diagnostic tools accounts for 10.3% of disability-adjusted life-years i Sunyer (IDIBAPS),
can assess an individuals’ susceptibility to ALD, and the (DALYs—a measure of overall disease burden that CIBER de
pathogenesis of this disease in humans is poorly under­ reflects the years of healthy life lost owing to both pre­ Enfermedades
Hepáticas y Digestivas
stood. This scenario is in marked contrast to the spec­ mature mortality and prolonged poor health), a figure (CIBERehd), Center
tacular leaps forward made in the treatment of other liver that comes second only to the morbidity associated with Esther Koplowitz,
Laboratory of Liver
diseases, such as hepatitis B and C. The lack of advances tobacco use in developed countries (11.7% of DALYs).9 Fibrosis, Rosellón 149,
in the field of ALD are mainly related to the difficulties Alcohol misuse also affects individuals in develop­ 3rd Floor, 08036
of conducting clinical trials in patients with an active ing countries, where it is responsible for almost 9% of Barcelona, Spain
(J. Altamirano,
alcohol addiction, the lack of interest in this field from DALYs.10,11 Worldwide, ALD is responsible for about 4% R. Bataller).
of DALYs.9
Correspondence to:
Competing interests ALD is probably the main cause of death among people R. Bataller
The authors declare no competing interests. with severe alcohol abuse and is responsible for about bataller@clinic.ub.es

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 8  |  SEPTEMBER 2011  |  491


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points
■■ Alcohol abuse is a major cause of preventable morbidity and mortality 80–90%
worldwide, and a major health problem in both the EU and the USA
■■ Alcoholic liver disease is a major cause of end-stage liver disease requiring liver
Healthy liver
transplantation
■■ The spectrum of alcoholic liver disease encompasses simple steatosis, Steatosis 20–40%
steatohepatitis, progressive fibrosis, cirrhosis and hepatocellular carcinoma
■■ The basic mechanisms of alcoholic liver disease have been evaluated in both
animal models and translational studies, but further research is needed to
confirm the results
■■ Studies in human liver samples have identified novel therapeutic targets for
alcoholic liver disease, including CXC chemokines, osteopontin, tumor necrosis
factor receptor superfamily member 12A receptor and nostrin
Alcoholic Fibrosis
■■ New experimental models of severe alcoholic liver disease that include 8–20%
steatohepatitis
profound hepatocellular damage and fibrosis should be developed

3.8% of global mortality.9 Moreover, ALD remains one 20–40%

of the most important causes of liver-related deaths in


the USA, with a mortality of 4.4 deaths per 100,000 of the
population—even higher than that of hepatitis C (2.9 Cirrhosis
3–10%
deaths per 100,000 people).12 Remarkably, data from the
European Liver Transplant Registry show that ALD was
recorded as the indication for liver transplantation in
almost half (41.6%) of procedures performed between
1996 and 2005.13,14 ALD is, therefore, the second most
common indication for liver transplantation in Europe
and North America.15,16
Clinical Hepatocellular
decompensations carcinoma
Natural history Figure 1 | The spectrum of alcoholic liver disease. The
ALD comprises a broad spectrum of disease: asymptom­ percentages represent the patients who progress from one
atic fatty liver, steatohepatitis, progressive fibrosis, stage to the next.
end-stage cirrhosis and superimposed hepatocellular
carcinoma (Figure 1). Up to 90% of patients with heavy
alcohol intake have some degree of steatosis,17 which is This model, therefore, enables the early identification of
usually asymptomatic and rapidly reversible with absti­ patients who do not respond to corticosteroid therapy
nence. Continued heavy alcohol consumption, however, and who could be considered for inclusion in clinical
leads to inflammation of the liver characterized by the trials (Figure 2).24
infiltration of polymorphonuclear leukocytes and hepato­ As alcohol abuse can lead to severe damage in the
cellular damage, both of which define alcoholic hepa­titis.18 nervous system, heart, kidney and pancreas, patients
Eventually, patients develop liver fibrosis deposition (20– with ALD may also develop a variety of symptoms unre­
40%) and cirrhosis (8–20%), which confers a high risk of lated to the liver, such as psychiatric manifestations, heart
complications (such as ascites, variceal bleeding, hepatic failure, renal failure and abdominal pain.
encephalo­pathy, renal failure and bacterial infections).19,20
Severe forms of alcoholic hepatitis have a very high short- Diagnosis and treatment
term mortality.20 The prognosis of patients with alcoholic The diagnosis of alcohol abuse is based on self-
hepatitis can be estimated by several predictive models, reporting, but collection of objective data is advisable.
such as Maddrey’s discriminant function, the model for Among the various biomarkers of chronic alcohol
end-stage liver disease (MELD), Glasgow scores and a abuse, carbohydrate-­d eficient transferrin (CDT) is
composite albumin, bilirubin, international normalized probably the most reliable.26 A combined index based
ratio and creatinine (ABIC) score (Figure 2).21–25 These on γ‑glutamyltransferase and CDT measurements
scores can help clinicians to manage these patients.21–25 has a good sensitivity for detecting excessive ethanol
In addition, response to corticosteroids could be evalu­ consumption.27
ated with the Lille model, which utilizes age, severity The cornerstone of management of patients with ALD
of renal impairment, albumin level, prothrombin time, is to stop them from drinking alcohol, as continued
initial bilirubin level, and change in bilirubin level after alcohol intake is the single most important risk factor
7 days of treatment: a value ≥0.45 identifies patients who for progression of ALD.28–30 Abstinence is also critical for
do not respond to corticosteroids and is associated with patients with advanced ALD, who may eventually require
a 25% probability of 6‑month survival, whereas a value liver transplantation, because patients who continue to
<0.45 reflects an 85% probability of 6‑month survival. drink alcohol are not eligible for most transplantation

492  |  SEPTEMBER 2011  |  VOLUME 8  www.nature.com/nrgastro


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

programs. Referral to rehabilitation programs, together Clinical and biochemical suspicion of alcoholic liver disease
with family support, is usually necessary to achieve absti­
nence in patients with ALD (Figure 3). Although studies
showed that drugs, such as disulfiram, naltrexone, acam­ Noninvasive diagnostic tests:
prosate and baclofen, can help some patients achieve sus­ ultrasonography, ASHtest, carbohydrate-deficient
transferrin, elastography
tained abstinence,31–34 drug treatment is not successful in versus
all patients. For those who find alcohol cessation impos­ Liver biopsy to rule out other etiologies

sible, treatments that attenuate the progression of liver


disease and reduce mortality are urgently needed.
Psychiatric evaluation
Parenteral and enteral feeding improve the nutritional
status of patients with alcoholic hepatitis without influ­
encing short-term survival.35 Several pharmacological
Group and supportive psychotherapy to
therapies have been tried in patients with severe alcoholic promote abstinence from alcohol
hepatitis, albeit with limited success (Figure 3). Studies
of anabolic–androgenic steroids36 and antioxidant drugs Failure Success
(such as vitamin E and silibinin37,38) showed that these Pharmacological therapy Consider an
agents did not improve survival. Other ineffective thera­ for alcoholism antifibrogenic therapy
(disulfiram, baclofen, naltrexone) (losartan, pentoxifylline)
pies include colchicine, propylthiouracil and combined
intravenous administration of glucagon and insulin.39–41 Figure 2 | Proposed algorithm for the treatment of
By contrast, treatment with corticosteroids (such as alcoholic hepatitis. The algorithm is based on current
evidence, AASLD Clinical Guidelines and experience in the
prednisolone) and pentoxifylline can prolong survival
Liver Unit of the Hospital Clinic of Barcelona.49 A value of
in some patients with alcoholic hepatitis, although nearly ≥0.45 in the Lille model is associated with a 25%
half of treated patients do not respond to this therapy.42–44 probability of survival for 6 months, whereas a value of
Corticosteroid treatment for patients with alcoholic <0.45 is associated with an 85% probability of 6‑month
hepa­titis, the most severe form of ALD, has been the survival.24 Abbreviations: ABIC, age, bilirubin, international
standard of care for the past 40 years. normalized ratio and creatinine; MARS, molecular
In addition, although some studies have investigated adsorbent recirculating system; MELD, model for end-stage
the potential role of agents that interfere with the bio­ liver disease.
logical effects of tumor necrosis factor (TNF), such
as the monoclonal antibody infliximab and the TNF
Clinical and biochemical suspicion of alcoholic hepatitis
inhibitor etanercept, in the treatment of patients with
severe alcoholic hepatitis, these two agents were associ­
ated with increased mortality owing to severe bacterial
Consider transjugular liver biopsy for histological confirmation
infections.45–48 As a result, novel targeted therapies are
needed for alcoholic hepatitis. Treatment algorithms for
ALD and alcoholic hepatitis based on current evidence,
Severity stratification
AASLD Clinical Guidelines and experience in the Liver (Maddrey’s discriminant function, MELD, ABIC and Glasgow scores)
Unit of the Hospital Clinic of Barcelona are proposed
(Figures 2 and 3).49
Intermediate and high risk Low risk
Disease progression
Steatosis
Alcoholic steatosis is a complex process manifested Severe infection No or Nutrition assessment and
controlled infection standard supportive care
through several mechanisms. The main pathogenetic
factors underlying this process are increased fatty acid
and triglyceride synthesis, enhanced hepatic influx of
Pentoxifylline Corticosteroids
free fatty acids from adipose tissue and of chylo­microns (400 mg three times (prednisone 40 mg)
daily for 4 weeks)
from the intestinal mucosa, increased hepatic lipo­ Lille model score at 7 days
genesis, inhibited lipolysis, and damaged mitochondria <0.45 ≥0.45
and microtubules, all of which result in accumula­tion of Stop corticosteroids
4 weeks of corticosteroids
VLDL.50–55 The molecular mechanisms involved in ste­ followed by a 2-week taper Consider for clinical trials
Consider for MARS
atosis are largely unknown, although overexpression of
lipogenic enzymes results from the down­regulation Figure 3 | Proposed algorithm for the treatment of alcoholic liver disease. The
of peroxisome proliferator activated receptor α (PPARα) algorithm is based on current evidence, AASLD Clinical Guidelines and experience
and the induction of sterol regulatory element-binding in the Liver Unit of the Hospital Clinic of Barcelona.49 The ASHtest (BioPredictive,
Paris, France) estimates the severity of alcoholic liver disease from the levels of
protein (SREBP).56,57 AMP-activated protein kinase,
α2-macroglobulin, haptoglobin, apolipoprotein A1, total bilirubin,
which regulates the relative concentrations of intra­ γ‑glutamyltransferase, alanine aminotransferase and aspartate aminotransferase.
cellular malonyl coenzyme A and long-chain acyl- Some parameters are adjusted for the patient’s age and sex. Abbreviations: ABIC,
­coenzyme A—the key metab­olites responsible for the age, bilirubin, international normalized ratio and creatinine; MARS, molecular
balance between fat synthesis and fat degradation adsorbent recirculating system; MELD, model for end-stage liver disease.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 8  |  SEPTEMBER 2011  |  493


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Continuous alcohol intake Various molecular mechanisms probably contribute


to the development of steatohepatitis (Figure 4). For
ADH, ALDH, CYP2E1
example, acetaldehyde (the major product of alcohol
metabolism) binds to both proteins and DNA, resulting
Gut permeability Antioxidants Acetaldehyde
in functional alterations and the formation of protein
Hepatocytes Kupffer cells adducts. In turn, these adducts form autoantigens that
Hepatic
stellate cells activate the adaptive immune system 64,65 and recruit
lympho­c ytes to the damaged liver. Acetaldehyde also
induces mitochondrial damage and impairs glutathione
Serum lipopolysaccharide ROS ROS function, leading to oxidative stress and apoptosis.66–68
Cytokines Oxidative stress, and the resulting protein nitrosyla­
CD14 Kupffer cells TGF-β
TLR4 Hepatic stellate cells tion and lipid peroxidation, are believed to have a
Lipid
major pathogenetic role in ALD.69 Nonparenchymal
NOx Mitochondrial
damage peroxidation liver cells are the major producers of reactive oxygen
species (ROS) in patients with ALD.70 The enzymes and
NFκB, JNK pathways involved in ROS generation include the cyto­
chrome P450 2E1 (CYP2E1)-dependent microsomal
electron transport system of the respiratory chain,
STATs Cytokines NADH-dependent cytochrome reductase and xanthine
oxidase.71–73 Moreover, chronic alcohol intake mark­
edly upregulates CYP2E1 activity, because this enzyme
Regeneration Inflammation Apoptosis Fibrosis (in addition to alcohol dehydrogenase and aldehyde
Figure 4 | Pathogenetic pathways leading to progression of alcoholic liver disease. dehydro­genase), metabolizes ethanol to acetaldehyde;
Ethanol promotes the translocation of lipopolysaccharide from the gastrointestinal thus alcohol ingestion stimulates the generation of ROS
lumen to the portal vein, where it binds to the lipopolysaccharide-binding protein. In and hydroxyl-ethyl radicals.70 Alcohol metabolites and
the Kupffer cells, lipopolysaccharide binds to CD14, which combines with TLR4 ROS stimulate signaling pathways that involve nuclear
activating multiple cytokine genes with inflammatory cytokine production and factor κB (NFκB), signal transducer and activator of
decreasing the expression of STATs leading to reduced liver regeneration. Long- transcription (STAT)–Janus kinase (JAK) and c‑Jun
term alcohol consumption alters the intracellular balance of antioxidants with
N‑terminal kinase (JNK) in hepatic resident cells, leading
subsequent decrease in the release of mitochondrial cytochrome c and expression
of Fas ligand, leading to hepatic apoptosis through the caspase‑3 activation to local synthesis of inflammatory mediators, such as
pathway. Studies in rats and mice suggest that activated Kupffer cells and tumor necrosis factor (TNF), IL‑17, CXC chemokines
hepatocytes are sources of free radicals (especially ROS), which are responsible (including IL‑8), as well as osteopontin.74–80
for lipid peroxidation and further apoptotic damage. Activation of hepatic stellate Alcohol abuse also results in changes in the colonic
cells also contributes to the production of cytokines, ROS and TGF‑β exacerbating microbiota and increased permeability of the intestine
liver fibrosis. Abbreviations: ADH, alcohol dehydrogenase; ALDH, aldehyde to bacterial endotoxin, which leads to elevated serum
dehydrogenase; CYP2E1, cytochrome P450 2E1; JNK, c‑Jun N‑terminal kinase;
levels of lipopolysaccharide. This increase in lipopoly­
NFκB, nuclear factor κB; NOx, nitrogen oxides; ROS, reactive oxygen species; STAT,
signal transducer and activator of transcription; TGF‑β, transforming growth
saccahride induces inflammatory actions in Kupffer cells
factor β; TLR4, Toll-like receptor 4. via the CD14–Toll-like receptor (TLR) 4 pathway.3,81 The
resulting inflammatory milieu in the alcoholic liver then
leads to polymorphonuclear leukocyte infiltration, ROS
pathways—is also implicated in hepatic fat metabolism.58 formation and hepatocellular damage. Finally, impaired
In addition, changes in NADH reduction–­oxidation protein degradation through the ubiquitin–proteasome
potential in the liver might promote lipogenesis pathway, increased endoplasmic reticulum stress and
through inhibition of fatty acid oxidation. Furthermore, altered autophagy also promote hepatocellular injury
microsomal tri­g lyceride transfer protein activity is and development of hepatic inclusions of aggregated
reduced by long-term ethanol administration, which cytokeratins (termed Mallory-Denk bodies).82
also causes fat accumula­tion in the liver.59
Fibrosis
Steatohepatitis Patients with alcoholic steatohepatitis can develop pro­
Steatohepatitis is defined by the presence of fatty liver, gressive fibrosis. In livers affected by ALD, the fibrotic
an inflammatory infiltrate (which mainly consists of tissue is typically located in pericentral and peri­
polymorphonuclear leukocytes) and hepatocellular sinusoidal areas.83 In advanced stages of ALD, colla­gen
damage. This stage of ALD is a prerequisite for progres­ bands are evident and bridging fibrosis develops, which
sion to fibrosis and cirrhosis.60 The histological subtypes precedes the development of regeneration nodules and
of steatohepatitis are not well defined and the severity of liver cirrhosis. The cellular and mol­ecular mechanisms of
disease might depend on the amount of alcohol intake fibrosis in ALD are not completely understood. Alcohol
and other environmental factors, such as diet and life­ metab­olites, such as acetaldehyde, can directly activate
style, as well as the genetic background of patient.61–63 hepatic stellate cells, the main collagen-­producing cells
When the inflammation and hepatocellular injury are in the injured liver.84 Hepatic stellate cells can also be
severe, the condition is called alcoholic hepatitis. activated in a paracrine fashion by damaged hepatocytes,

494  |  SEPTEMBER 2011  |  VOLUME 8  www.nature.com/nrgastro


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

activated Kupffer cells and polymorphonuclear leuko­ focus of investigation. Genetic factors that influence the
cytes.85 The damaged hepatocytes, activated Kupffer cells activity of these enzymes and the rate of alcohol metab­
and polymorphonuclear leukocytes release a wide range olism have also been extensively studied. Owing to its
of fibrogenic mediators: growth factors, such as trans­ fibrogenic potential, variations in the rate of genera­
forming growth factor (TGF)‑β and platelet-derived tion of acetaldehyde could explain the differences in the
growth factor; cytokines, including leptin, angiotensin II, suscepti­bility of individuals to ALD after abusive alcohol
IL‑8 and TNF; nitric oxide; and ROS (super­oxide, consumption. Most genetic epidemiological studies of
hydrogen peroxide and hydroxyl anion radicals). 86,87 ALD have been performed in Asia, where the preva­
Importantly, ROS stimulate profibrogenic intracellular lence of polymorphisms in the genes encoding enzymes
signaling pathways in hepatic stellate cells, including involved in alcohol metabolism, such as ADH1B, ADH1C
extracellular signal-­regulated kinases (ERK1 and ERK2), and ALDH2, is very high. Although polymorphisms in
phosphoinositide 3 kinase–Akt and JNK.88,89 ROS also these genes and in the 5'-flanking region of CYP2E1 are
upregulate tissue inhibitor of metallo­proteinases 1 accepted to be involved in an individual’s susceptibility to
and decrease the action of metallo­proteinases, which alcoholism, their role in the progression of ALD remains
jointly promote collagen accumulation.90,91 Cells other controversial.99 A meta-analysis of studies on the associa­
than hepatic stellate cells, including portal fibroblasts tion between alcoholism and alcohol-induced liver
and bone-marrow-derived cells, can also synthesize damage highlighted the heterogeneity of the case–control
collagen in patients with ALD. Whether other novel studies that investigated ADH1B, ADH1C, CYP2E1 and
mechanisms, such as epi­thelial-to-mesenchymal trans­ ADLH2 polymorphisms.100
ition of hepatocytes, also have a role in liver fibrosis is Polymorphisms in genes that encode pro­inflammatory
under investigation.92 cytokines involved in the pathogenesis of ALD have
also been examined. Although a single nucleotide
Environmental and genetic factors poly­morphism located in the promoter region of TNF
ALD results from a complex interaction of behavioral, (which increases production of this cytokine) is associ­
environmental and genetic factors. Although a posi­ ated with severe steatohepatitis, no studies have yet
tive correlation between cumulative alcohol intake and confirmed this finding. Polymorphisms in the genes
degrees of liver fibrosis has been reported, extensive encoding NFκB subunits, IL‑1β and IL‑1 receptor
variability in the histological response to alcohol abuse antagonists, IL‑2, IL‑6 and IL‑10 also modify the pro­
exists in individuals.93,94 At similar levels of ethanol con­ gression of ALD.98 Moreover, studies have also investi­
sumption, some patients only develop macrovesicular gated the role of genetic variation in factors involved
steatosis, while others develop progressive fibrosis and in lipopolysaccharide-­induced intracellular pathways,
cirrhosis. The main environmental factors that promote including CD14 and TLR4, as potential risk factors for
the progression of liver fibrosis in patients with alcohol ALD. Differential expression of genes belonging to these
abuse include obesity and cigarette smoking. 87,95 The pathways could alter the inflammatory and fibrogenic
role of metabolic factors, including insulin resistance, is response of the liver after prolonged alcohol intake.101
unclear, although some reports suggest that such factors Patients with polymorphisms that result in low activity
might also favor progression of fibrosis.96 of superoxide dismutase and gluthatione‑S-transferase—
Genetic factors are also known to influence the organ two powerful antioxidant enzymes—are also at risk of
specificity of alcohol-related harmful effects. While developing severe ALD. Finally, three studies indicated
some patients with heavy alcohol consumption develop that variations in PNPLA3, which encodes patatin-like
ALD, others mainly suffer from alcohol-related dis­ phospholipase domain-­containing protein 3, strongly
orders of the pancreas, heart and nervous system. influence the development of advanced liver fibrosis
Studies in families showed variations in the suscepti­ among Mexican and Caucasian populations. 61,102,103
bility to ALD between different ethnic groups, suggest­ Importantly, PNPLA3 polymorphisms can be consid­
ing that genetic factors are important determinants of ered to be the only confirmed and replicated genetic risk
disease risk. Twin studies also suggest a major role for factor for ALD.
genetic pre­d isposition in the susceptibility to ALD; Despite the large number of studies that have assessed
concordance rates for alcoholism and alcohol-induced the role of genetic variation in susceptibility to ALD, a
liver fibrosis in monozygotic twins were 26.3 and 14.6, large-scale, well-designed, genome-wide association
respectively. 97 Epidemiological studies suggest that study of factors associated with ALD remains to be
several genetic factors influence the severity of steatosis performed. Most published reports include a limited
and oxidative stress, and that the cytokine milieu, the number of patients who lack a well-defined phenotype,
magnitude of the immune response, and/or the severity have only investigated a few genes and have important
of fibrosis also modulate an indivi­duals’ propensity to methodological weaknesses. Consequently, a genetic
progress to advanced ALD.98 test capable of identifying patients with the propensity
Genes encoding the main alcohol-metabolizing to develop advanced ALD has not yet been developed.
enzymes (alcohol dehydrogenase, aldehyde dehydrogenase Such a genetic test could be useful in clinical settings,
and CYP2E1) and proteins involved in the toxic effects as it would help to identify the main genetic determi­
of alcohol and its metabolites on the liver, such as anti­ nants of ALD, which would assist in the development of
oxidants and proinflammatory cytokines, have been the new therapies.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 8  |  SEPTEMBER 2011  |  495


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Potential therapeutic targets for alcoholic liver disease*


Target molecule Biological function Disease
IL‑8 and GRO‑α75,76 Inflammation, hepatocellular death Alcoholic hepatitis
NFκB, TLR3 and TLR7141 Fibrogenesis, inflammation, hepatocellular death Alcoholic liver disease (various stages)
Osteopontin 77
Inflammation, hepatocellular death Alcoholic liver disease (various stages)
IL‑17142 Inflammation, hepatocellular death Alcoholic hepatitis
P90SRK 143
Fibrogenesis, hepatocellular death Alcoholic hepatitis
TNF receptor superfamily Fibrogenesis, inflammation, hepatocellular death Alcoholic liver disease (various stages)
member 12A107
ADAMTS13 and von Fibrinolysis, coagulation Alcoholic hepatitis
Willebrand factor111,112
Nostrin113 Endoplasmic reticulum stress Alcoholic cirrhosis
Dimethylarginines 144,145
Endoplasmic reticulum stress, endothelial dysfunction Alcoholic hepatitis
IL‑22 and STAT379,106 Inflammation, hepatocellular death Alcoholic hepatitis
Fas108 Hepatocellular death, cellular apoptosis Alcoholic hepatitis
Bcl‑2109
Hepatocellular death, cellular apoptosis Alcoholic hepatitis
IL‑6146 Inflammation Alcoholic liver disease (various stages)
TNF105 Inflammation Alcoholic hepatitis
*All were identified in in vitro studies that used human liver samples. Abbreviations: ADAMTS13, a disintegrin and metalloproteinase with a thrombospondin
type 1 motif, member 13; GRO‑α, growth-regulated α‑protein; P90SRK, p90 ribosomal S6 kinase; TNF, tumor necrosis factor.

New therapeutic targets in humans CXC chemokines: IL‑18 and GRO‑α


As animal models do not accurately mimic the main find­ This family of chemokines mainly attracts polymorpho­
ings in advanced ALD in humans, studies using human nuclear leukocytes, the main infiltrating inflammatory
tissue are needed to identify new therapeutic targets. As cells in ALD. In patients with alcoholic hepatitis, the
discussed above, epidemiological studies have revealed baseline levels of expression of some of these chemokines
several genes that mediate injury and fibrosis in ALD. in the liver correlated with the patient’s survival duration
However, the key genes involved in the pathogenesis and the degree of portal hypertension.75,76
of ALD are unclear. Studies have also investigated the
correla­tion between serum levels of molecular media­ IL‑22–STAT3 pathway
tors of ALD (such as TNF) and disease severity,104 but IL‑22 has been identified as a member of the IL‑10 family
the pathogenetic significance of the associations identi­ of cytokines, which are produced by T helper type 17 cells
fied in these studies is similarly unclear, since increased and natural killer cells. This interleukin has important
serum levels of cyto­kines could result from impaired roles in the control of bacterial infection, homeo­stasis
liver clearance or ongoing bacterial infections. A more and tissue repair.106 Treatment with IL‑22 ameliorates
rational approach would be to investigate the expression experimental ALD in rodent models through activa­
and/or activation of different mediators of ALD in liver tion of STAT3. Moreover, IL‑22 receptor 1 expression is
tissue from patients with ALD, and to correlate these upregulated, whereas IL‑22 expression is undetectable,
findings with disease severity and the patient’s outcome. in patients with alcoholic hepatitis.79
This approach has already been used to identify poten­
tial targets for therapy in patients with severe ALD TNF superfamily receptors
(Table 1). However, the pathogenesis of ALD involves In patients with alcoholic hepatitis, TNF is not over­
interactions between multiple genes and environmental expressed. By contrast, several members of the TNF
factors. The results of gene-array studies that identified receptor superfamily are markedly upregulated. In par­
single genetic determinants of ALD should, therefore, be ticular, TNF receptor superfamily member 12A (also
considered cautiously.75,105 known as Fn14 or the Tweak receptor) is markedly over­
A wide variety of cytokines, immune modulators and expressed in these patients and its expression correlates
markers of fibrogenesis are altered in patients with ALD. with the severity of alcoholic hepatitis.107 Interestingly,
However, the fact that their tissue expression or serum TNFRSF12A is mainly expressed in hepatic progenitor
levels are altered in this setting might not render these cells, which accumulate in patients with severe forms of
specific mediators useful as therapeutic targets. Although alcoholic hepatitis.
some of these molecules take part in the physiopathol­
ogy of ALD, others might be more useful as indicators Osteopontin
of disease activity and inflammation than for their role The extracellular matrix protein osteopontin is highly
as disease mediators. The most relevant mediators for expressed in patients with alcoholic hepatitis. The basal
ALD identified in human samples are discussed in level of expression of osteopontin correlates with the
detail below. severity of this disease and its expression is upregulated

496  |  SEPTEMBER 2011  |  VOLUME 8  www.nature.com/nrgastro


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 2 | The most important experimental models of alcoholic liver disease


Outline of model Species Liver damage Advantages Disadvantages References
Direct ethanol administration Rats, Steatosis Controlled ethanol dose Lack of utility as a model of 110–113
via upper gastrointestinal tract mice, Mild polymorphonuclear chronic ethanol administration
intubation (esophageal or rabbits leukocyte infiltration Stressful
gastric) Not physiological
Tsukamoto–French model of Rats, mice Severe steatosis Good control of ethanol Expensive maintenance 141–144
ethanol administration via Scattered hepatocyte necrosis intake Surgical expertise needed
permanent intragastric Mild polymorphonuclear Possibility of high blood Not suitable for acute
gastrostomy leukocyte and mononuclear ethanol levels exposure studies
infiltration
Lieber–De Carli liquid nutrient Rats, Steatosis No bias resulting from Important differences between 145, 146
diet: ethanol accounts for mice, Mild inflammation nonalcoholic nutrient animal and human pathology
36%* of total calories baboons, Baboons: alcoholic hepatitis intake, owing to exactly Baboons: high cost and
Control littermates receive minipigs features and cirrhosis equivalent caloric intake methodologically difficult
carbohydrates as substitute Minipigs: resembles the by littermate controls Minipigs: high cost and lack of
histological features of human long-term results
alcoholic liver disease
Combination of Lieber–De Carli Rats, mice Marked steatosis No bias resulting from Important differences between 114, 115
diet and jejunoileal bypass, or Focal polymorphonuclear nonalcoholic nutrient animal and human pathology
combination of Lieber–De Carli leukocyte infiltration intake, owing to exactly Methodologically difficult
diet plus endotoxin Occasional hepatocyte necrosis equivalent caloric intake Not physiological
administration by littermate controls
Knockout and transgenic Mice Depends on the function of the Enables specific and Findings are difficult to 120–123
animals manipulated gene systematic analysis of translate to human diseases
Mostly affects macrovesicular genes and their products, Not physiological
and microvesicular fat as well as their relation Methodologically difficult
deposition to ethanol administration
Voluntary oral ethanol intake: a Rats Marked steatosis Resembles human Important differences between 124
diet containing 8% ethanol, fish Focal polymorphonuclear drinking habits animal and human pathology
oil (30% of calories), protein leukocyte infiltration Possibility of high blood
and dextrose is administered Occasional hepatocyte necrosis ethanol levels and
for 8 weeks Pericentral liver collagen considerable liver injury,
deposition including mild fibrosis
Chronic–binge ethanol intake: Mice Marked steatosis Resembles human Methodologically difficult 119
a 5% liquid ethanol diet is Focal polymorphonuclear drinking habits Force-feeding (gavage)
administered for 10 days, leukocyte infiltration Possibility of high blood procedure requires technical
followed by a force-fed single Focal hepatocyte necrosis ethanol levels and expertise
dose of 20% ethanol (5 g/kg considerable liver injury
body weight)
*In the minipig nutrient diet ethanol accounts for 40% of total calories.

by fibrogenic mediators, such as TGF‑β. Importantly, which result in microcirculatory disturbances. In


mice that lack osteopontin develop attenuated liver patients with alcoholic hepatitis, the imbalance between
injury after prolonged exposure to alcohol.77 ADAMTS13 activity and these unusually large von
Willebrand factor multimers caused by proinflammatory
Proapoptotic molecules: Fas and Bcl‑2 cytokines could contribute to sinusoidal micro­circulatory
Several studies have demonstrated that proapoptotic disturbances and subsequent liver injury.111,112
mediators, such as Fas and Bcl‑2, are upregulated in the
livers of patients with ALD.108,109 Oxidative stress stimu­ Nostrin
lates the expression of these proteins, and thereby pro­ Nostrin, also known as nitric oxide synthase traffic
motes the induction of apoptotic pathways. Although inducer, regulates nitric oxide (NO) synthesis and is a key
these mediators are appealing targets for the treatment effector in chronic liver diseases. A marked increase in
of patients with ALD, prolonged inhibition of their the expression of nostrin protein and mRNA is evident
expression could favor cancer development, which is in the livers of patients with ALD, which could contrib­
already a major complication in patients with chronic ute to the decreased enzymatic activity of endothelial NO
liver diseases.110 synthase in diseased livers.113

ADAMTS13: von Willebrand factor cleaving protease Animal models of ALD


Decreased plasma activity of ADAMTS13 (a disintegrin A number of strategies have been proposed to induce
and metalloproteinase with a thrombospondin type 1 ALD in rodents and other animals by continuous expo­
motif, member 13), which is produced by hepatic stel­ sure to ethanol or ethanol-containing diets (Table 2).114–119
late cells, leads to accumulation of unusually large von The first limitation of this approach is the reluctance of
Willebrand factor multimers and subsequent thrombi, most rodents to drink ethanol-containing diets. Only

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 8  |  SEPTEMBER 2011  |  497


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

some genetically selected rats (Marchigian Sardinan incidence of cirrhosis seen in patients with ALD.79 This
alcohol-preferring rats) will spontaneously ingest ethanol model could, therefore, closely reproduce the drinking
at high concentrations and display a binge-like drinking behaviors of humans.
behavior.120 Moreover, most studies are usually designed New animal models that mimic the pathological find­
to induce ALD within few weeks, and consequently the ings induced by ethanol exposure in humans are clearly
extent of liver disease is modest. needed. Future studies will need to test whether, when
Administration of ethanol in these animal models combined with excessive alcohol intake, other factors,
ranges from voluntary oral intake via drinking water such as obesity and cigarette smoking, participate in the
or a liquid diet (Lieber–De Carli model) to direct pathogenesis of ALD. Additionally, studying the effects
gastric administration via a nasogastric tube or perm­ of ethanol exposure in animal models of comorbidities
anent gastro­stomy (Tsukamoto–French model).121–124 are required to investigate the observed synergy between
Experiments using these models are usually performed alcohol abuse and other factors that promote liver
under controlled nutritional conditions. Solutions of disease (such as an adipogenic diet and viral hepatitis)
5–20% ethanol are used, and very high blood alcohol and to study the increased liver oncogenesis caused by
levels can be achieved. Hepatic steatosis, a mild ethanol abuse.
inflamma­tory infiltrate and modest liver injury develop
after 4–12 weeks of alcohol administration. However, the Conclusions
main histological findings in patients with severe ALD Excessive alcohol consumption is a major preventable
(massive fibrosis and hepatocellular damage) are not cause of morbidity and mortality worldwide, and ALD is
always present, which limits the value of these models. a frequent indication for liver transplantation. However,
An additional limitation is the rapid circadian changes in despite its high prevalence, ALD has received little atten­
ethanol metabolism in rodents, which should be closely tion in the past two decades and no major advance has
monitored to avoid acute intoxication. been made in the management of these patients since
The histological findings in baboons and minipigs the introduction of corticosteroid therapy. The develop­
fed with ethanol-containing diets closely mimic those of ment of targeted therapies for ALD is hampered by poor
ALD, with sequential development of fatty liver disease, knowledge of the molecular mechanisms involved in its
alcoholic steatohepatitis and cirrhosis (Table 2). 125–127 development, particularly in humans, and by the percep­
However, none of the currently used animal models of tion that it is an addictive and a self-inflicting disease.
ALD results in portal hypertension, which is a major Most studies of the mechanisms underlying ALD have
determinant of mortality in patients with severe ALD. been conducted in animal models, which do not repro­
A widely used strategy to increase the patho­ duce all the pathological changes found in humans with
genic effects of ethanol in rodents is simultaneous severe forms of the disease.
administra­tion of other agents known to contribute to In addition to these animal studies, however, trans­
its deleterious effect on the liver. Intraperitoneal injec­ lational research using tissue samples from patients
tions of lipo­p olysaccharide and supplementation of with ALD have identified novel potential therapeutic
iron in the diet both increase liver inflammation and targets for this disease. However, most of the data so
injury.128–130 Similarly, a diet containing high levels of far obtained are preliminary and the roles of the identi­
poly­unsaturated fatty acids exacerbates fatty liver and fied molecules will need to be characterized in trans­
resultant hepatic injury after ethanol exposure. The data genic and other animal models. In the future, studies of
from these studies should be considered with caution, biological systems that integrate the existing informa­
however, as the adjuvant agents administered in conjunc­ tion at different levels (such as genetic, metabolic and
tion with ethanol can themselves induce patho­logical proteomic data) will be required to delineate the pre­
effects in the liver. vailing pathogenetic pathways in ALD. The potential
The role of specific genes in the development of ALD value of drugs that target these molecules will need
has been investigated using genetically manipulated to be initially evaluated in preclinical settings, before
mice (transgenic models). These genes include Ppar, clinical implementation.
Rxr, Srebf1, Cyp2e1, Tnfrsf1a and Tnfrsf1b, Ncf1 (which
encodes p47-phox), Cd14, Tlr4, Icam1, Serpine1, IL6 and
Nfe2l2, among others.131–139 For instance, transgenic mice
overexpressing Cyp2e1 that were given an alcohol-rich
diet had significantly higher serum alanine transaminase Review criteria
levels and showed higher histological injury to liver than
A search for original articles was performed in PubMed
wild-type mice.140 using the following terms: “alcoholism”, “alcoholic liver
Another mouse model that combines chronic alcohol disease”, “steatohepatitis” and “alcoholic hepatitis”, in
exposure and binge ethanol administration has been pro­ combination with “incidence”, “prevalence”, “therapy”,
posed. Binge drinking is the most common pattern of “translational”, “experimental models”, “classification”,
alcohol consumption in adolescents. Although animal “prognosis”, “diagnosis” and “treatment”. Data from
models of ethanol binges might not completely mimic full-text articles and abstracts published in the English
the liver injury observed in humans, some studies indi­ language were reviewed. Relevant articles were also
identified from the reference lists of included papers.
cate a substantial contribution of binge drinking to the

498  |  SEPTEMBER 2011  |  VOLUME 8  www.nature.com/nrgastro


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

1. Becker, U. et al. Prediction of risk of liver disease 23. Dominguez, M. et al. A new scoring system for 42. O’Shea, R. S., Dasarathy, S. & McCullough, A. J.
by alcohol intake, sex, and age: a prospective prognostic stratification of patients with Alcoholic liver disease. Am. J. Gastroenterol.
population study. Hepatology 23, 1025–1029 alcoholic hepatitis. Am. J. Gastroenterol. 103, 105, 14–32 (2010).
(1996). 2747–2756 (2008). 43. Akriviadis, E. et al. Pentoxifylline improves short-
2. Tome, S. & Lucey, M. R. Review article: 24. Louvet, A. et al. The Lille model: a new tool for term survival in severe acute alcoholic hepatitis:
current management of alcoholic liver disease. therapeutic strategy in patients with severe a double-blind, placebo-controlled trial.
Aliment. Pharmacol. Ther. 19, 707–714 (2004). alcoholic hepatitis treated with steroids. Gastroenterology 119, 1637–1648 (2000).
3. Gao, B. et al. Innate immunity in alcoholic liver Hepatology 45, 1348–1354 (2007). 44. Mathurin, P. et al. Corticosteroids improve short-
disease. Am. J. Physiol. Gastrointest. Liver 25. Forrest, E. H. et al. Analysis of factors predictive term survival in patients with severe alcoholic
Physiol. 300, G516–G525 (2011). of mortality in alcoholic hepatitis and derivation hepatitis: meta-analysis of individual patient
4. Goodman, Z., Makhlouf, H. & Younossi, Z. Reply. and validation of the Glasgow alcoholic hepatitis data. Gut 60, 255–260 (2011).
Hepatology 54, 370–371 (2011). score. Gut 54, 1174–1179 (2005). 45. Naveau, S. et al. A double-blind randomized
5. Gleeson, D. et al. HFE genotypes in 26. Bortolotti, F., De Paoli, G. & Tagliaro, F. controlled trial of infliximab associated with
decompensated alcoholic liver disease: Carbohydrate-deficient transferrin (CDT) as a prednisolone in acute alcoholic hepatitis.
phenotypic expression and comparison with marker of alcohol abuse: a critical review of the Hepatology 39, 1390–1397 (2004).
heavy drinking and with normal controls. Am. J. literature 2001–2005. J. Chromatogr. B Analyt. 46. Spahr, L. et al. Combination of steroids with
Gastroenterol. 101, 304–310 (2006). Technol. Biomed. Life Sci. 841, 96–109 (2006). infliximab or placebo in severe alcoholic
6. Singal, A. K., Sagi, S., Kuo, Y. F. & Weinman, S. 27. Hietala, J. et al. Comparison of the combined hepatitis: a randomized controlled pilot study.
Impact of hepatitis C virus infection on the marker GGT–CDT and the conventional J. Hepatol. 37, 448–455 (2002).
course and outcome of patients with acute laboratory markers of alcohol abuse in heavy 47. Tilg, H. et al. Anti-tumor necrosis factor-α
alcoholic hepatitis. Eur. J. Gastroenterol. Hepatol. drinkers, moderate drinkers and abstainers. monoclonal antibody therapy in severe alcoholic
23, 204–209 (2011). Alcohol Alcohol. 41, 528–533 (2006). hepatitis. J. Hepatol. 38, 419–425 (2003).
7. Gitto, S., Micco, L., Conti, F., Andreone, P. & 28. Pessione, F. et al. Five-year survival predictive 48. Menon, K. V. et al. A pilot study of the safety and
Bernardi, M. Alcohol and viral hepatitis: a mini- factors in patients with excessive alcohol intake tolerability of etanercept in patients with
review. Dig. Liver Dis. 41, 67–70 (2009). and cirrhosis. Effect of alcoholic hepatitis, alcoholic hepatitis. Am. J. Gastroenterol. 99,
8. Clouston, A. D., Jonsson, J. R. & Powell, E. E. smoking and abstinence. Liver Int. 23, 45–53 255–260 (2004).
Steatosis as a cofactor in other liver diseases: (2003). 49. O’Shea, R. S., Dasarathy, S. & McCullough, A. J.,
hepatitis C virus, alcohol, hemochromatosis, and 29. Chedid, A. et al. Prognostic factors in alcoholic Practice Guideline Committee of the American
others. Clin. Liver Dis. 11, 173–189 (2007). liver disease. VA Cooperative Study Group. Am. Association for the Study of Liver Diseases &
9. The world health report (2002)—reducing risks, J. Gastroenterol. 86, 210–216 (1991). Practice Parameters Committee of the American
promoting healthy life. World Health Organization 30. Amini, M. & Runyon, B. A. Alcoholic hepatitis College of Gastroenterology. Alcoholic liver
[online], http://www.who.int/whr/2002/en/ 2010: a clinician’s guide to diagnosis and disease. Hepatology 51, 307–328 (2010).
(2002). therapy. World J. Gastroenterol. 16, 4905–4912 50. Salaspuro, M. P., Shaw, S., Jayatilleke, E.,
10. Cortez-Pinto, H. et al. The burden of disease and (2010). Ross, W. A. & Lieber, C. S. Attenuation of the
the cost of illness attributable to alcohol 31. Addolorato, G. et al. Effectiveness and safety of ethanol‑induced hepatic redox change after
drinking-results of a national study. Alcohol. Clin. baclofen for maintenance of alcohol abstinence chronic alcohol consumption in baboons:
Exp. Res. 34, 1442–1449 (2010). in alcohol- dependent patients with liver metabolic consequences in vivo and in vitro.
11. Rehm, J. et al. Global burden of disease and cirrhosis: randomised, double-blind controlled Hepatology 1, 33–38 (1981).
injury and economic cost attributable to alcohol study. Lancet 370, 1915–1922 (2007). 51. Sozio, M. S., Liangpunsakul, S. & Crabb, D. The
use and alcohol-use disorders. Lancet 373, 32. Palmer, A. J. et al. The long-term cost- role of lipid metabolism in the pathogenesis of
2223–2233 (2009). effectiveness of improving alcohol abstinence alcoholic and nonalcoholic hepatic steatosis.
12. Paula, H. et al. Alcoholic liver disease-related with adjuvant acamprosate. Alcohol Alcohol. 35, Semin. Liver Dis. 30, 378–390 (2010).
mortality in the United States: 1980–2003. Am. 478–492 (2000). 52. Zhou, J. et al. Hepatic fatty acid transporter
J. Gastroenterol. 105, 1782–1787 (2010). 33. Garbutt, J. C. et al. Efficacy and tolerability of CD36 is a common target of, LXR, PXR, and
13. European liver transplant registry. ELTR.org long-acting injectable naltrexone for alcohol PPARγ in promoting steatosis. Gastroenterology
[online], http://www.eltr.org/spip.php?spip0 dependence: a randomized controlled trial. JAMA 134, 556–567 (2008).
(2011). 293, 1617–1625 (2005). 53. Coll, O., Colell, A., Garcia-Ruiz, C., Kaplowitz, N.
14. Mackie, J. et al. Orthotopic liver transplantation 34. Fuller, R. K. & Gordis, E. Does disulfiram have a & Fernández-Checa. J. C. Sensitivity of the
for alcoholic liver disease: a retrospective role in alcoholism treatment today? Addiction 2-oxoglutarate carrier to alcohol intake
analysis of survival, recidivism, and risk factors 99, 21–24 (2004). contributes to mitochondrial glutathione
predisposing to recidivism. Liver Transplant. 7, 35. Stickel, F., Hoehn, B., Schuppan, D. & Seitz, H. K. depletion. Hepatology 38, 692–702 (2003).
418–427 (2001). Nutritional therapy in alcoholic liver disease. 54. Hoek, J. B., Cahill, A. & Pastorino, J. G. Alcohol
15. Transplant fast facts. Arbor Research Aliment. Pharmacol. Ther. 18, 357–373 (2003). and mitochondria: a dysfunctional relationship.
Collaborative for Health [online], http:// 36. Mendenhall, C. L. et al. Short-term and long-term Gastroenterology 122, 2049–2063 (2002).
www.ustransplant.org (2011). survival in patients with alcoholic hepatitis 55. Sozio, M. & Crabb, D. W. Alcohol and lipid
16. Burra, P. et al. Liver transplantation for alcoholic treated with oxandrolone and prednisolone. metabolism. Am. J. Physiol. Endocrinol. Metab.
liver disease in Europe: a study from the ELTR N. Engl. J. Med. 311, 1464–1470 (1984). 295, E10–E16 (2008).
(European Liver Transplant Registry). Am. J. 37. Mezey, E., Potter, J. J., Rennie-Tankersley, L., 56. Galli, A., Pinaire, J., Fischer, M., Dorris, R. &
Transplant. 10, 138–148 (2010). Caballeria, J. & Pares, A. A randomized placebo Crabb, D. W. The transcriptional and DNA binding
17. Lefkowitch, J. H. Morphology of alcoholic liver controlled trial of vitamin E for alcoholic activity of peroxisome proliferator-activated
disease. Clin. Liver Dis. 9, 37–53 (2005). hepatitis. J. Hepatol. 40, 40–46 (2004). receptor α is inhibited by ethanol metabolism.
18. Elphick, D. A., Dube, A. K., McFarlane, E., 38. Parés, A. et al. Effects of silymarin in alcoholic A novel mechanism for the development of
Jones, J. & Gleeson, D. Spectrum of liver patients with cirrhosis of the liver: results of a ethanol-induced fatty liver. J. Biol. Chem. 276,
histology in presumed decompensated alcoholic controlled, double-blind, randomized and 68–75 (2001).
liver disease. Am. J. Gastroenterol. 102, multicentre trial. J. Hepatol. 28, 615–621 57. You, M., Fischer, M., Deeg, M. A. & Crabb, D. W.
780–788 (2007). (1998). Ethanol induces fatty acid synthesis pathways by
19. Adachi, M. & Brenner, D. A. Clinical syndromes of 39. Rambaldi, A. & Gluud, C. Meta-analysis of activation of sterol regulatory element-binding
alcoholic liver disease. Dig. Dis. 23, 255–263 propylthiouracil for alcoholic liver disease—a protein (SREBP). J. Biol. Chem. 277,
(2005). Cochrane Hepato-Biliary Group Review. Liver 21, 29342–29347 (2002).
20. Lucey, M. R., Mathurin, P. & Morgan, T. R. 398–404 (2001). 58. You, M., Matsumoto, M., Pacold, C. M.,
Alcoholic hepatitis. N. Engl. J. Med. 360, 40. Akriviadis, E. A. et al. Failure of colchicine to Cho, W. K. & Crabb, D. W. The role of AMP-
2758–2769 (2009). improve short-term survival in patients with activated protein kinase in the action of ethanol
21. Maddrey, W. C. et al. Corticosteroid therapy of alcoholic hepatitis. Gastroenterology 99, in the liver. Gastroenterology 127, 1798–1808
alcoholic hepatitis. Gastroenterology 75, 811–818 (1990). (2004).
193–199 (1978). 41. Trinchet, J. C. et al. Treatment of severe alcoholic 59. Sugimoto, T. et al. Decreased microsomal
22. Dunn, W. et al. MELD accurately predicts hepatitis by infusion of insulin and glucagon: triglyceride transfer protein activity contributes
mortality in patients with alcoholic hepatitis. a multicenter sequential trial. Hepatology 15, to initiation of alcoholic liver steatosis in rats.
Hepatology 41, 353–358 (2005). 76–81 (1992). J. Hepatol. 36, 157–162 (2002).

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 8  |  SEPTEMBER 2011  |  499


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

60. Bataller, R., Rombouts, K., Altamirano, J. & 79. Ki, S. H. et al. Interleukin-22 treatment 100. Zintzaras, E., Stefanidis, I., Santos, M. & Vidal, F.
Marra, F. Fibrosis in alcoholic and nonalcoholic ameliorates alcoholic liver injury in a murine Do alcohol-metabolizing enzyme gene
steatohepatitis. Best Pract. Res. Clin. model of chronic-binge ethanol feeding: role of polymorphisms increase the risk of alcoholism
Gastroenterol. 25, 231–244 (2011). signal transducer and activator of transcription 3. and alcoholic liver disease? Hepatology 43,
61. Tian, C., Stokowski, R. P., Kershenobich, D., Hepatology 52, 1291–1300 (2010). 352–361 (2006).
Ballinger, D. G. & Hinds, D. A. Variant in PNPLA3 80. Lemmers, A. et al. The interleukin-17 pathway is 101. Jarvelainen, H. A. et al. Promoter polymorphism
is associated with alcoholic liver disease. Nat. involved in human alcoholic liver disease. of the CD14 endotoxin receptor gene as a risk
Genet. 42, 21–23 (2010). Hepatology 49, 646–657 (2009). factor for alcoholic liver disease. Hepatology 33,
62. Altamirano, J. et al. The amount of alcohol 81. Yin, M. et al. Reduced early alcohol-induced liver 1148–1153 (2001).
consumption negatively impacts short-term injury in CD14-deficient mice. J. Immunol. 166, 102. Stickel, F. et al. Genetic variation in the PNPLA3
mortality in patients with alcoholic hepatitis. 4737–4742 (2001). gene is associated with alcoholic liver injury in
Am. J. Gastroenterol. doi:10.1038/ 82. Donohue, T. M. Jr, Cederbaum, A. I. & caucasians. Hepatology 53, 86–95 (2011).
ajg.2011.141. French, S. W. Role of the proteasome in ethanol- 103. Trépo, E. et al. Common polymorphism in the
63. MacSween, R. N. & Burt, A. D. Histologic induced liver pathology. Alcohol. Clin. Exp. Res. PNPLA3/adiponutrin gene confers higher risk of
spectrum of alcoholic liver disease. Semin. Liver 31, 1446–1459 (2007). cirrhosis and liver damage in alcoholic liver
Dis. 6, 221–232 (1986). 83. Yip, W. W. & Burt, A. D. Alcoholic liver disease. disease. J. Hepatol. doi:10.1016/
64. Svegliati-Baroni, G., Baraona, E., Rosman, A. S. Semin. Diagn. Pathol. 23, 149–160 (2006). j.jhep.2011.01.028.
& Lieber, C. S. Collagen–acetaldehyde adducts 84. Friedman, S. L. Stellate cell activation in 104. Bird, G. L. et al. Increased plasma tumor
in alcoholic and nonalcoholicliver diseases. alcoholic fibrosis—an overview. Alcohol. Clin. Exp. necrosis factor in severe alcoholic hepatitis.
Hepatology 20, 111–118 (1994). Res. 23, 904–910 (1999). Ann. Intern. Med. 112, 917–920 (1990).
65. You, M. & Crabb, D. W. Recent advances in 85. Wang, J. H., Batey, R. G. & George, J. Role of 105. Domίnguez, M. et al. Functional analysis of
alcoholic liver disease II. Minireview: ethanol in the regulation of hepatic stellate cell hepatic gene expression profiling from patients
molecular mechanisms of alcoholic fatty liver. function. World J. Gastroenterol. 12, 6926–6932 with alcoholic hepatitis reveals new targets for
Am. J. Physiol. Gastrointest. Liver Physiol. 287, (2006). therapy. J. Hepatol. 50 (Suppl. 1), S39 (2009).
G1–G6 (2004). 86. Moreno, M. & Bataller, R. Cytokines and renin– 106. Horiguchi, N. et al. Cell type-dependent pro- and
66. Zhou, Z., Sun, X. & Kang, Y. J. Ethanol-induced angiotensin system signaling in hepatic fibrosis. anti-inflammatory role of signal transducer and
apoptosis in mouse liver: Fas- and cytochrome Clin. Liver Dis. 12, 825–852 (2008). activator of transcription 3 in alcoholic liver injury.
c-mediated caspase-3 activation pathway. Am. J. 87. Naveau, S. et al. Serum leptin in patients with Gastroenterology 134, 1148–1158 (2008).
Pathol. 159, 329–338 (2001). alcoholic liver disease. Alcohol. Clin. Exp. Res. 30, 107. Domίnguez, M. et al. Identification of Fn14, a
67. Bradham, C. A., Plümpe, J., Manns, M. P., 1422–1428 (2006). TNF receptor superfamily member, as a
Brenner, D. A. & Trautwein, C. Mechanisms of 88. Kluwe, J. et al. Modulation of hepatic fibrosis by therapeutic target in patients with alcoholic
hepatic toxicity. I. TNF-induced liver injury. Am. J. c‑Jun‑N-terminal kinase inhibition. hepatitis. Hepatology 50 (Suppl. 4), 609A
Physiol. 275, G387–G392 (1998). Gastroenterology 138, 347–359 (2010). (2009).
68. Román, J. et al. Differential role of ethanol and 89. Houstis, N., Rosen, E. D. & Lander, E. S. Reactive 108. Ribeiro, P. S. et al. Hepatocyte apoptosis,
acetaldehyde in the induction of oxidative stress oxygen species have a causal role in multiple expression of death receptors, and activation of
in HEP G2 cells: effect on transcription factors forms of insulin resistance. Nature 440, NF-κB in the liver of nonalcoholic and alcoholic
AP-1 and NF-κB. Hepatology 30, 1473–1480 944–948 (2006). steatohepatitis patients. Am. J. Gastroenterol.
(1999). 90. Lieber, C. S. et al. Value of fibrosis markers for 99, 1708–1717 (2004).
69. Meagher, E. A. et al. Alcohol-induced generation staging liver fibrosis in patients with precirrhotic 109. Ramalho, R. M. et al. Apoptosis and Bcl-2
of lipid peroxidation products in humans. J. Clin. alcoholic liver disease. Alcohol. Clin. Exp. Res. 32, expression in the livers of patients with
Invest. 104, 805–813 (1999). 1031–1039 (2008). steatohepatitis. Eur. J. Gastroenterol. Hepatol.
70. Bailey, S. M. & Cunningham, C. C. Acute and 91. Arthur, M. J., Iredale, J. P. & Mann, D. A. Tissue 18, 21–29 (2006).
chronic ethanol increases reactive oxygen inhibitors of metalloproteinases: role in liver 110. Kalaitzakis, E., Gunnarsdottir, S. A.,
species generation and decreases viability in fibrosis and alcoholic liver disease. Alcohol. Clin. Josefsson, A. & Björnsson, E. Increased risk for
fresh, isolated rat hepatocytes. Hepatology 28, Exp. Res. 23, 940–943 (1999). malignant neoplasms among patients with
1318–1328 (1998). 92. Zhu, L. et al. Hepatic deletion of Smad7 in mouse cirrhosis. Clin. Gastroenterol. Hepatol. 9,
71. Mansouri, A. et al. An alcoholic binge causes leads to spontaneous liver dysfunction and 168–174 (2011).
massive degradation of hepatic mitochondrial aggravates alcoholic liver injury. PLoS ONE 6, 111. Ishikawa, M. et al. Potential role of enhanced
DNA in mice. Gastroenterology 117, 181–190 e17415 (2011). cytokinemia and plasma inhibitor on the
(1999). 93. Weber, S. N. & Lammert, F. Genetics of liver injury decreased activity of plasma ADAMTS13 in
72. Ji, C. & Kaplowitz, N. Betaine decreases and fibrosis. Alcohol. Clin. Exp. Res. 35, 800–803 patients with alcoholic hepatitis: relationship to
hyperhomocysteinemia, endoplasmic reticulum (2011). endotoxemia. Alcohol. Clin. Exp. Res.
stress, and liver injury in alcohol-fed mice. 94. Wilfred de Alwis, N. M. & Day, C. P. Genetics of 34 (Suppl. 1), S25–S33 (2010).
Gastroenterology 124, 1488–1499 (2003). alcoholic liver disease and nonalcoholic fatty liver 112. Matsuyama, T. et al. Increased von Willebrand
73. Yin, M. et al. Alcohol-induced free radicals in disease. Semin. Liver Dis. 27, 44–54 (2007). factor over decreased ADAMTS13 activity may
mice: direct toxicants or signaling molecules? 95. Altamirano, J. & Bataller, R. Cigarette smoking contribute to the development of liver
Hepatology 34, 935–942 (2001). and chronic liver diseases. Gut 59, 1159–1162 disturbance and multiorgan failure in patients
74. Bird, G. L., Sheron, N., Goka, A. K., (2010). with alcoholic hepatitis. Alcohol. Clin. Exp. Res.
Alexander, G. J. & Williams, R. S. Increased 96. Stepanova, M., Rafiq, N. & Younossi, Z. M. 31 (Suppl. 1), S27–S35 (2007).
plasma tumor necrosis factor in severe Components of metabolic syndrome are 113. Mookerjee, R. P. et al. Increased gene and
alcoholic hepatitis. Ann. Intern. Med. 112, independent predictors of mortality in patients protein expression of the novel eNOS regulatory
917–920 (1990). with chronic liver disease: a population-based protein NOSTRIN and a variant in alcoholic
75. Dominguez, M. et al. Hepatic expression of CXC study. Gut 59, 1410–1415 (2010). hepatitis. Gastroenterology 132, 2533–2541
chemokines predicts portal hypertension and 97. Hrubec, Z. & Omenn, G. S. Evidence of genetic (2007).
survival in patients with alcoholic hepatitis. predisposition to alcoholic cirrhosis and 114. Bor, S. et al. Effect of ethanol on the structure
Gastroenterology 136, 1639–1650 (2009). psychosis: twin concordances for alcoholism and and function of rabbit esophageal epithelium.
76. Colmenero, J. et al. Hepatic expression of its biological end points by zygosity among male Am. J. Physiol. 274, G819–G826 (1998).
candidate genes in patients with alcoholic veterans. Alcohol. Clin. Exp. Res. 5, 207–215 115. Bor, S., Bor-Caymaz, C., Tobey, N. A.,
hepatitis: correlation with disease severity. (1981). Abdulnour-Nakhoul, S. & Orlando, R. C.
Gastroenterology 132, 687–697 (2007). 98. Bataller, R., North, K. E. & Brenner, D. A. Genetic Esophageal exposure to ethanol increases risk
77. Morales, O. et al. Osteopontin is a novel polymorphisms and the progression of liver of acid damage in rabbit esophagus. Dig. Dis.
therapeutic target in patients with alcoholic fibrosis: a critical appraisal. Hepatology 37, Sci. 44, 290–300 (1999).
hepatitis. J. Hepatol. 52 (Suppl. 1), S23 (2010). 493–503 (2003). 116. Salo, J. A. Ethanol-induced mucosal injury in
78. Horiguchi, N. et al. Cell type-dependent pro- and 99. Day, C. P. et al. Investigation of the role of rabbit oesophagus. Scand. J. Gastroenterol. 18,
anti-inflammatory role of signal transducer and polymorphisms at the alcohol and aldehyde 713–721 (1983).
activator of transcription 3 in alcoholic liver dehydrogenase loci in genetic predisposition to 117. Yamada, Y., Weller, R. O., Kleihues P &
injury. Gastroenterology 134, 1148–1158 alcohol-related end-organ damage. Hepatology Ludeke, B. I. Effects of ethanol and various
(2008). 14, 798–801 (1991). alcoholic beverages on the formation of

500  |  SEPTEMBER 2011  |  VOLUME 8  www.nature.com/nrgastro


© 2011 Macmillan Publishers Limited. All rights reserved
REVIEWS

O6-methyldeoxyguanosine from concurrently coadministration of endotoxin and ethanol on rat 139. Lamlé, J. et al. Nuclear factor-eythroid 2-related
administered N-nitrosomethylbenzylamine in liver pathology and proinflammatory and anti- factor 2 prevents alcohol-induced fulminant
rats: a dose-–response study. Carcinogenesis inflammatory cytokines. Hepatology 29, liver injury. Gastroenterology 134, 1159–1168
13, 1171–1175 (1992). 1503–1510 (1999). (2008).
118. Tsukamoto, H., Towner, S. J., Yu, G. S. & 129. Bode, C., Gast, J., Zelder, O., Jerusalem, C. R. & 140. Morgan, K., French, S. W. & Morgan, T. R.
French, S. W. Potentiation of ethanol-induced Bode, J. C. Alcohol-induced liver injuries after Production of a cytochrome P450 2E1
pancreatic injury by dietary fat. Induction of jejunoileal bypass operation in rats. J. Hepatol. transgenic mouse and initial evaluation of
chronic pancreatitis by alcohol in rats. Am. J. 5, 75–84 (1987). alcoholic liver damage. Hepatology 36,
Pathol. 131, 246–257 (1988). 130. Zhang, H. Y. et al. Multiple pathogenic factor- 122–134 (2002).
119. Tsukamoto, H., Reidelberger, R. D., French, S. W. induced complications of cirrhosis in rats: 141. Stärkel, P., De Saeger, C., Strain, A. J.,
& Largman, C. Long-term cannulation model for a new model of hepatopulmonary syndrome Leclercq, I. & Horsmans, Y. NFκB, cytokines,
blood sampling and intragastric infusion in the with intestinal endotoxemia. World J. TLR 3 and 7 expression in human end-stage
rat. Am. J. Physiol. 247, R595–R599 (1984). Gastroenterol. 13, 3500–3507 (2007). HCV and alcoholic liver disease. Eur. J. Clin.
120. Ciccocioppo, R. et al. Genetically selected 131. Yin, M. et al. Essential role of tumor necrosis Invest. 40, 575–584 (2010).
Marchigian Sardinian alcohol-preferring (msP) factor α in alcohol-induced liver injury in mice. 142. Lemmers, A. et al. The interleukin-17 pathway
rats: an animal model to study the neurobiology Gastroenterology 117, 942–952 (1999). is involved in human alcoholic liver disease.
of alcoholism. Addict. Biol. 11, 339–355 (2006). 132. Kono, H. et al. CYP2E1 is not involved in early Hepatology 49, 646–657 (2009).
121. Tsukamoto, H. et al. Severe and progressive alcohol-induced liver injury. Am. J. Physiol. 277, 143. Morales-Ibanez, O. et al. Proteomic analysis
steatosis and focal necrosis in rat liver induced G1259–G1267 (1999). identifies P90RSK as a novel therapeutic
by continuous intragastric infusion of ethanol 133. Kono, H. et al. NADPH oxidase-derived free target in patients with alcoholic hepatitis.
and low fat diet. Hepatology 5, 224–232 (1985). radicals are key oxidants in alcohol‑induced J. Hepatol. 54 (Suppl. 1), S9 (2011).
122. French, S. W. Intragastric ethanol infusion model liver disease. J. Clin. Invest. 106, 867–872 144. Mookerjee, R. P. et al. Increasing
for cellular and molecular studies of alcoholic (2000). dimethylarginine levels are associated with
liver disease. J. Biomed. Sci. 8, 20–27 (2001). 134. Tsukamoto, H. et al. Experimental liver cirrhosis adverse clinical outcome in severe alcoholic
123. Lieber, C. S. & DeCarli, L. M. An experimental induced by alcohol and iron. J. Clin. Invest. 96, hepatitis. Hepatology 45, 62–71 (2007).
model of alcohol feeding and liver injury in the 620–630 (1995). 145. Lluch, P. et al. Plasma concentrations of nitric
baboon. J. Med. Primatol. 3, 153–163 (1974). 135. Crabb, D. W., Galli, A., Fischer, M. & You, M. oxide and asymmetric dimethylarginine in
124. Lieber, C. S., Jones, D. P. & DeCarli, L. M. Effects Molecular mechanisms of alcoholic fatty liver: human alcoholic cirrhosis. J. Hepatol. 41,
of prolonged ethanol intake: production of fatty role of peroxisome proliferator-activated 55–59 (2004).
liver despite adequate diets. J. Clin. Invest. 44, receptor α. Alcohol 34, 35–38 (2004). 146. Naveau, S. et al. Harmful effect of adipose
1009–1021 (1965). 136. Miller, A. M. et al. Inflammation-associated tissue on liver lesions in patients with
125. Lieber, C. S., DeCarli, L. & Rubin, E. Sequential IL-6/STAT3 activation ameliorates alcoholic and alcoholic liver disease. J. Hepatol. 52,
production of fatty liver, hepatitis, and cirrhosis nonalcoholic fatty liver diseases in IL-10 895–902 (2010).
in sub-human primates fed ethanol with deficient mice. Hepatology doi:10.1002/
adequate diets, Proc. Natl Acad. Sci. USA 72, hep.24517. Acknowledgments
437–441 (1975). 137. Kono, H. et al. ICAM-1 is involved in the The authors of this article are supported by grants
126. Niemelä, O. et al. Sequential acetaldehyde mechanism of alcohol-induced liver injury: from Fondo de Investigación Sanitaria (FIS PI080237
production, lipid peroxidation, and fibrogenesis studies with knockout mice. Am. J. Physiol. and FIS PS09/01164). J. Altamirano also holds a
in micropig model of alcohol-induced liver Gastrointest. Liver Physiol. 280, G1289–G1295 grant from Fundación Banco Bilbao Vizcaya Argentaria
disease. Hepatology 22, 1208–1214 (1995). (2001). (FBBVA). CIBERehd is funded by the Instituto de Salud
127. Halsted, C. H. et al. Centrilobular distribution of 138. You, M., Matsumoto, M., Pacold, C. M., Carlos III.
acetaldehyde and collagen in the ethanol-fed Cho, W. K. & Crabb, D. W. The role of AMP-
micropig. Hepatology 18, 954–960 (1993). activated protein kinase in the action of ethanol Author contributions
128. Järveläinen, H. A., Fang, C., Ingelman- in the liver. Gastroenterology 127, 1798–1808 Both authors contributed equally to all aspects of this
Sundberg, M. & Lindros K. O. Effect of chronic (2004). Review.

NATURE REVIEWS | GASTROENTEROLOGY & HEPATOLOGY VOLUME 8  |  SEPTEMBER 2011  |  501


© 2011 Macmillan Publishers Limited. All rights reserved

Das könnte Ihnen auch gefallen