Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Alcohol 34 (2004) 9–19

Alcoholic fatty liver: its pathogenesis and mechanism of


progression to inflammation and fibrosis
Charles S. Lieber*
Bronx Veterans Affairs Medical Center, Bronx, NY 10468, USA
Mount Sinai School of Medicine, New York, NY 10029, USA
Received 30 March 2004; received in revised form 16 July 2004; accepted 20 July 2004

Abstract
Liver disease in the alcoholic is due not only to malnutrition but also to ethanol’s hepatotoxicity linked to its metabolism by means of
the alcohol dehydrogenase and cytochrome P450 2E1 (CYP2E1) pathways and the resulting production of toxic acetaldehyde. In addition,
alcohol dehydrogenase–mediated ethanol metabolism generates the reduced form of nicotinamide adenine dinucleotide (NADH), which
promotes steatosis by stimulating the synthesis of fatty acids and opposing their oxidation. Steatosis is also promoted by excess dietary
lipids and can be attenuated by their replacement with medium-chain triglycerides. Through reduction of pyruvate, elevated NADH also
increases lactate, which stimulates collagen synthesis in myofibroblasts. Furthermore, CYP2E1 activity is inducible by its substrates, not
only ethanol but also fatty acids. Their excess and metabolism by means of this pathway generate release of free radicals, which cause oxidative
stress, with peroxidation of lipids and membrane damage, including altered enzyme activities. Products of lipid peroxidation such as
4-hydroxynonenal stimulate collagen generation and fibrosis, which are further increased through diminished feedback inhibition of collagen
synthesis because acetaldehyde forms adducts with the carboxyl-terminal propeptide of procollagen in hepatic stellate cells. Acetaldehyde
is also toxic to the mitochondria, and it aggravates their oxidative stress by binding to reduced glutathione and promoting its
leakage. Oxidative stress and associated cellular injury promote inflammation, which is aggravated by increased production of the
proinflammatory cytokine tumor necrosis factor-alpha in the Kupffer cells. These are activated by induction of their CYP2E1 as well as
by endotoxin. The endotoxin-stimulated tumor necrosis factor-alpha release is decreased by dilinoleoylphosphatidylcholine, the active
phosphatidylcholine (PC) species of polyenylphosphatidylcholine (PPC). Moreover, defense mechanisms provided by peroxisome proliferator-
activated receptor alpha and omega fatty acid oxidation are readily overwhelmed, particularly in female rats and also in women who
have low hepatic induction of fatty acid–binding protein (L-FABPc). Accordingly, the intracellular concentration of free fatty acids may
become high enough to injure membranes, thereby contributing to necrosis, inflammation, and progression to fibrosis and cirrhosis. Even-
tually, hepatic S-adenosylmethionine and PCs become depleted in the alcoholic, with impairment of their multiple cellular functions, which
can be restored by PC replenishment. Thus, prevention and therapy opposing the development of steatosis and its progression to more
severe injury can be achieved by a multifactorial approach: control of alcohol consumption, avoidance of obesity and of excess dietary
long-chain fatty acids, or their replacement with medium-chain fatty acids, and replenishment of S-adenosylmethionine and PCs by using
PPC. Progress in the understanding of the pathogenesis of alcoholic fatty liver and its progression to inflammation and fibrosis has resulted
in prospects for their better prevention and treatment. 쑖 2005 Elsevier Inc. All rights reserved.
Keywords: Alcoholic steatosis; Alcoholic steatohepatitis; Cytochrome P450 2E1; Obesity; Dilinoleoylphosphatidylcholine; S-adenosylmethionine; Oxidative
stress; TNF-α; Acetaldehyde

1. Introduction dominant metabolism in the liver associated with oxidation–


reduction (redox) changes and oxidative stress. Redox
Progress in the understanding of the pathogenesis of alco-
changes are mediated by alcohol dehydrogenase (ADH), and
holic liver disease was achieved when it was discovered
oxidative stress is generated mainly by the activity of the
that alcohol affects the liver through not only nutritional
microsomal ethanol oxidizing system (MEOS) and its key
disturbances but also its direct toxicity because of its pre-
enzyme cytochrome P450 2E1 (CYP2E1), which releases
free radicals.
* Corresponding author. Bronx Veterans Affairs Medical Center, 130 The clinical course and ultimate outcome of alcoholic
West Kingsbridge Road (151-2), Bronx, NY 10468, USA. Tel.: ⫹1-718-
741-4244; fax: ⫹1-718-733-6257.
liver disease are dismal. In a prospective survey of 280
E-mail address: liebercs@aol.com (C.S. Lieber). patients with alcoholic liver injury, Chedid et al. (1991)
Editor: T.R. Jerrells found that, within 48 months of follow-up, 30% of those
0741-8329/05/$ – see front matter 쑖 2005 Elsevier Inc. All rights reserved.
doi: 10.1016/j.alcohol.2004.07.008
10 C.S. Lieber / Alcohol 34 (2004) 9–19

with a fatty liver, more than half of those with cirrhosis, and This, however, did not prevent the development of the first
two-thirds of those with cirrhosis plus alcoholic hepatitis stage of alcoholic liver disease, namely alcoholic fatty liver.
died. This outcome is more severe than that of many Therefore, the question was raised whether alcohol may
cancers, yet it is attracting much less concern, among both have some direct toxic effect on the liver independent of
the public and the medical profession. This may be due, at malnutrition (Fig. 1). This hypothesis was initially rejected
least in part, to the prevailing and pervasive perception because, as stated by Best et al. (1949, p. 1005), “there is
that not much can be done about this major public health no more evidence of a specific toxic effect of pure ethyl
issue. Pathogenic concepts of alcoholic liver disease, how- alcohol upon liver cells than there is for one due to sugar”.
ever, are evolving, and elucidation of the biochemical effects This was based on experiments in rats given adequate diets
of ethanol allows for a more optimistic outlook in terms of with alcohol in drinking water. They did not develop any
diagnosis and treatment. liver damage unless the diet was made deficient. However,
rats do not drink enough alcohol when given in drinking
water for significant blood concentrations to develop. Their
2. Pathogenesis of alcoholic and non-alcoholic fatty liver aversion was overcome by incorporating the added ethanol
into totally liquid, nutritionally adequate diets containing
2.1. Alcohol and nutrition
35% of calories as alcohol. Under those conditions, an obvi-
Unlike other drugs, ethanol is a substantial source of ous fatty liver developed compared with the findings in
energy, with 7.1 kcal (29.7 kJ) per gram, a value that exceeds control animals pair-fed with an isocaloric diet in which
the energy content of carbohydrates or proteins. On average, alcohol was replaced by carbohydrates (Lieber et al., 1963).
ethanol accounts for half an alcoholic’s caloric intake. It Similar results were obtained in human volunteers (Lieber
therefore displaces normal nutrients, causing malnutrition et al., 1963, 1965). Results of these studies clearly estab-
(Fig. 1), including deficiencies of folate, thiamine, and other lished that, even in the presence of an adequate diet, alcohol
vitamins. Secondary malnutrition also occurs through malab- can produce a fatty liver and at blood concentrations that
sorption due to gastrointestinal complications, such as pan- do not necessarily produce intoxication. Increases in die-
creatic insufficiency and impaired hepatic metabolism of tary long-chain triglycerides enhanced the alcohol-induced
nutrients (Fig. 1). In addition, alcohol promotes the degrada- steatosis, and this effect was attenuated by medium-chain
tion of nutrients, as exemplified by its effects on vitamin A triglycerides (Lieber et al., 1967). Decreasing of dietary fat
(Lieber, 1992). reduced steatosis; however, below a concentration of 10%
Because, experimentally, nutritional deficiencies cause of calories as dietary fat, there was a reversal of this effect,
liver damage, it was postulated that this is also the mecha- probably owing to the high carbohydrate content of the
nism whereby alcoholic liver disease develops. As a result, corresponding diet (Lieber & DeCarli, 1970). These effects
the prevailing therapy was nutritional with enriched diets. of alcohol were shown to be due to its metabolism in the liver.

Fig. 1. Interaction of the direct toxicity of ethanol with malnutrition resulting from primary or secondary deficiencies. Secondary malnutrition may be
caused by either (1) maldigestion and malabsorption or (2) impaired utilization due to decreased activation or increased degradation of nutrients. Both
direct toxicity of ethanol and malnutrition (whether primary or secondary) may affect function and structure of the liver and gastrointestinal tract.
C.S. Lieber / Alcohol 34 (2004) 9–19 11

2.2. Toxic effects of hepatic alcohol oxidation 2.3. Microsomal ethanol oxidizing system
Oxidation of ethanol through the ADH pathway produces As reviewed elsewhere (Lieber, 1997, 1999), the discov-
acetaldehyde, which is converted to acetate. Both reac- ery of the MEOS was prompted by the observation that, after
tions reduce nicotinamide adenine dinucleotide (NAD) to chronic ethanol consumption, there is an adaptive increase of
its reduced form (NADH) (Fig. 2). Excess NADH causes a ethanol metabolism, which could not be explained on the
number of metabolic disorders (Lieber, 1992), including basis of ADH, therefore raising the possibility of the exis-
inhibition of the Krebs cycle and of its fatty acid oxidation. tence of an additional pathway. The first indication of a
The inhibition of fatty acid oxidation favors steatosis and possible interaction of ethanol with the endoplasmic reticu-
hyperlipidemia. The effects of ethanol were reproduced in lum of the hepatocyte (also called microsomal fraction when
vitro by an alternative NADH-generating system (sorbitol– obtained by ultracentrifugation) was provided by the obser-
fructose) and were blocked by an H⫹ acceptor (methylene vation that, in rats and human beings, ethanol feeding results
blue) (Lieber et al., 1959; Lieber & Schmid, 1961). The in a proliferation of the smooth endoplasmic reticulum (SER)
preventive effect of methylene blue against ethanol-induced (Iseri et al., 1964, 1966; Lane & Lieber, 1966), which was
fat accumulation was confirmed subsequently (Galli et al., confirmed by an increase in the amount of enzyme activities
1999). This rat model of alcoholic liver disease consistently of the hepatic microsomal membranes (Ishii et al., 1973a),
reproduced a fatty liver, but it did not progress to inflam- including a rise in microsomal glucose-6-phosphatase activ-
mation, fibrosis, and cirrhosis. This is probably due to the ity after chronic ethanol administration (Ishii et al., 1973b).
fact that, even with the liquid diet, the alcohol intake is This increase in SER resembled that seen after the adminis-
limited to 35% of total calories, whereas alcoholics in whom tration of a wide variety of hepatotoxins (Meldolesi, 1967),
cirrhosis develops usually consume at least 50% of their barbiturates, and other therapeutic agents (Conney, 1967) and
calories as alcohol. Such a high intake of alcohol was subse- food additives (Lane & Lieber, 1967). Because many of the
quently achieved in non-human primates, namely baboons substances that induce a proliferation of the SER are metab-
(Lieber & DeCarli, 1974a). In that animal model, it was also olized, at least in part, by the cytochrome P450 enzyme
shown that, in addition to the metabolic abnormalities caused system that is located in the SER, the possibility that ethanol
by the ADH activity, a new pathway of ethanol metabolism, may also be metabolized by a similar process was raised.
namely MEOS, plays a key role in the progression of the Indeed, such a system, named MEOS, was demonstrated in
disease. liver microsomes in vitro and found to be inducible by

Fig. 2. Ethanol oxidation by (A) alcohol dehydrogenase (ADH) and nicotinamide adenine dinucleotide (NAD⫹); (B) the hepatic microsomal ethanol
oxidizing system [(MEOS), which involves cytochrome P450 2E1 and the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH)];
(C) a combination of NADPH oxidase and catalase; and (D) xanthine oxidase and catalase.
12 C.S. Lieber / Alcohol 34 (2004) 9–19

chronic ethanol feeding in vivo (Lieber & DeCarli, 1968, the term MEOS characterizes total microsomal ethanol
1970). The proposal that this new pathway plays a significant oxidation, not only that catalyzed by CYP2E1.
role in ethanol metabolism initiated a decade of lively debate: The rat CYP2E1 gene was isolated, characterized, and
Some invoked that ADH was contaminating the liver micro- localized to chromosome 7 (Umeno et al., 1988b), and the
somes (Isselbacher & Carter, 1970), whereas others asserted human gene was isolated, characterized, and localized to
that this microsomal ethanol oxidation was due to a hydrogen chromosome 10 (Umeno et al., 1988a). In rabbits, two genes
peroxide–dependent reaction promoted by contaminating may be involved (Khani et al., 1988).
catalase (Ziegler, 1972). Indeed, it had been postulated that Enhanced concentrations of both hepatic CYP2E1 pro-
the combination of hydrogen peroxide generation from the tein and mRNA were found in actively drinking patients
reduced form of nicotinamide adenine dinucleotide phos- (Takahashi et al., 1993). As revealed from results of the
phate (NADPH) oxidase and of catalase could account for studies cited above and reviewed elsewhere (Lieber, 1997,
microsomal ethanol oxidation (Roach et al., 1969; Tephly 1999), the most common cause for CYP2E1 induction is
et al., 1969) (Fig. 1), especially because a hydrogen perox- early alcoholic liver injury, such as alcoholic steatosis and
ide–generating system (glucose–glucose oxidase) can be alcoholic steatohepatitis. Cytochrome P450 2E1, however,
substituted for NADPH. Actually, the latter is not unex- is also induced in nonalcoholic steatohepatitis, which can be
pected, because not only do microsomes contain catalase, understood on the basis of the physiologic role of CYP2E1.
but commercial glucose oxidase (used to generate NADPH) Indeed, there seems to be a dual role of CYP2E1 (Fig. 3),
is contaminated with catalase. It also had been reported that namely one of detoxification and one of nutritional support.
microsomes obtained from acatalasemic mice fail to oxidize That CYP2E1 contributes to the defense mechanisms of the
ethanol (Vatsis & Schulman, 1973), but this claim was body against the penetration of toxic xenobiotics is suggested
subsequently retracted (Vatsis & Schulman, 1974). Hepatic by its location and inducibility at port of entries into the body,
microsomes of acatalasemic mice subjected to heat inactiva- and by its broad substrate specificity.
tion displayed decreased catalase activity, but NADPH- In terms of its nutritional role, CYP2E1 is inducible by
dependent MEOS remained active and unaffected (Lieber & fasting in the rat (Koop & Tierney, 1990). The increase
DeCarli, 1974b; Teschke et al., 1975). Eventually, MEOS may be due, at least in part, to ketones. Indeed, in rats
was solubilized and separated from ADH and catalase activi- (Casazza et al., 1984), rabbits (Koop & Casazza, 1985), and
ties by diethylaminoethyl cellulose column chromatography human beings (Reichard et al., 1986), acetone seems to
(Teschke et al., 1972, 1975). be actively used, being metabolized by a microsomal acetone
It was established that the key enzyme of the MEOS monooxygenase identified as CYP2E1 (Casazza & Veech,
is the ethanol-inducible CYP2E1, which was found to be 1985; Johansson et al., 1986). Acetone is both an inducer
increased fourfold to 10-fold in liver biopsy samples obtained and a substrate of CYP2E1 (Koop, 1992; Yang et al., 1990).
from recently drinking subjects (Tsutsumi et al., 1989), Cytochrome P450 2E1 was also found to be induced in
with a corresponding rise in mRNA (Takahashi et al., obese rats (Raucy et al., 1991). The role of CYP2E1 in fatty
1993). This induction contributes to the metabolic tolerance acid metabolism supports the concept of a nutritional role
to ethanol that develops in the alcoholic (in addition to its for CYP2E1. Indeed, in addition to its ethanol-oxidizing
CNS tolerance).
Finally, successful reconstitution of the system was ac-
complished with either partially purified or highly purified
microsomal P-450 obtained from alcohol-treated (Ohnishi &
Lieber, 1977) or phenobarbital-treated (Miwa et al., 1978)
rats. In a new nomenclature system, it was proposed that
the ethanol-inducible form be designated as CYP2El (Nelson
et al., 1993). On the basis of findings of these and various
other studies, and regardless of the original claim to the
contrary (Thurman et al., 1972), it was finally agreed by
the principal contenders involved that catalase cannot ac-
count for the microsomal ethanol oxidation (Teschke et al.,
1977; Thurman & Brentzel, 1977), and that the MEOS is Fig. 3. Physiologic and toxic roles of cytochrome P450 2E1 (CYP2E1),
the main cytochrome P450 of the microsomal ethanol oxidizing system
distinct from ADH and catalase and dependent on cyto- (MEOS). Many endogenous and xenobiotic compounds, including ethanol,
chromes P450, as reviewed elsewhere (Lieber, 1997, 1999). ketones, and fatty acids, are substrates for CYP2E1 and induce its activity
Despite the discovery of CYP2E1 and its prevailing role through various mechanisms (see text), resulting in an array of beneficial as
in microsomal ethanol oxidation, the term MEOS was main- well as harmful effects. NADPH ⫽ Reduced form of nicotinamide adenine
dinucleotide phosphate. Reprinted from C. S. Lieber, Microsomal ethanol-
tained because cytochromes P450 other than CYP2E1 (such oxidizing system (MEOS): the first 30 years (1968–1998)—a review,
as CYP1A2 and CYP3A4) (Salmela et al., 1998) may also Alcoholism: Clinical and Experimental Research 23(6), 991–1007, 1999,
contribute to ethanol metabolism in the microsomes. Thus, with permission of Lippincott, Williams & Wilkins.
C.S. Lieber / Alcohol 34 (2004) 9–19 13

activity, CYP2E1 catalyzes fatty acid ω-1 and ω-2 hydroxyl- supra and Fig. 3); their excess up-regulates CYP2E1. Al-
ations (Adas et al., 1998; Amet et al., 1994; Laethem though the pathogenesis of non-alcoholic fatty liver disease
et al., 1993). Ethanol feeding also results in an increased and nonalcoholic steatohepatitis has not yet been fully eluci-
activity of CYP4A1 (Ma et al., 1993). The CYP4A subfam- dated, a popular mechanism is the “two hit” theory (James &
ily catalyzes ω-hydroxylation at the terminal carbon of fatty Day, 1998), with the first hit being the accumulation, by
acids. Further oxidation of the ω and ω-1 hydroxyacids several causes (e.g., obesity), of fatty acids in the liver.
by alcohol and ADH results in the production of dicarboxylic The second hit is the peroxidation of these fatty acids because
and oxocarboxylic acids. The oxidation of ω-hydroxyacids of the oxidative stress produced by different factors such as
is principally catalyzed by class 3 ADH, which has a low CYP2E1 induction (Angulo & Lindor, 2001).
affinity for ethanol, whereas the ω-l hydroxyacids are likely The damage caused by oxidative stress in both alcoholic
to be preferentially oxidized by class 1 ADH, which has a steatohepatitis and nonalcoholic steatohepatitis includes mi-
high affinity for ethanol (Boleda et al., 1993). tochondrial injury, which, in turn, exacerbates the oxidative
It is of interest that dicarboxylic acids (products of the stress (Lieber, 1997). That mitochondrial damage is a key
CYP4A-mediated pathway) play a regulatory role in the component of alcoholic liver injury is well established
hepatic disposition of nonesterified fatty acids by activating (Lieber, 1992). However, nonalcoholic steatohepatitis is also
the peroxisome proliferator-activated receptor alpha, which associated with mitochondrial structural defects, whereas
increases the transcription of key fatty acid–disposal path- non-alcoholic fatty liver disease is not. This mitchondrial
ways, such as the microsomal CYP4A1, the peroxisomal dysfunction contributes to the oxidative stress in nonalco-
acyl-coenzyme A oxidase, and the liver fatty acid–binding holic steatohepatitis (Sanyal et al., 2001).
protein in the cytosol (L-FABPc) (Kaikaus et al., 1993). Like many other useful adaptive systems, when the adap-
This, in turn, results in enhanced microsomal omega-oxida- tation becomes excessive, adverse consequences prevail.
tion and peroxisomal beta-oxidation of fatty acids and in Cytochrome P450 2E1 leaks oxygen radicals as part of its
a greater stimulatory effect of L-FABPc on microsomal acyl- operation (Fig. 3), and when they exceed the cellular defense
glycerol synthesis (Burnett et al., 1979). systems they result in oxidative stress with its pathologic
The resulting changes exacerbate the sex difference in consequences. This is true when excess alcohol has to be
the alcohol-induced lipid abnormalities and in the vulnera- metabolized, as in alcoholic steatohepatitis, or when
CYP2E1 is confronted by an excess of ketones and fatty
bility to alcoholic liver disease. It was known that sex dif-
acids associated with diabetes, obesity, or both, resulting in
ferences in ethanol distribution (Jones & Jones, 1976),
nonalcoholic steatohepatitis.
bioavailability (Frezza et al., 1990a), and hepatic metabo-
lism (Mishra et al., 1989), with increased production of acet-
aldehyde (Eriksson et al., 1996), may contribute to women’s 3. Progression of fatty liver to inflammation and fibrosis
vulnerability to alcohol consumption. In addition, sex differ-
The oxidative stress caused by CYP2E1 induction and
ences in alcohol-induced derangements of hepatic lipid
mitochondrial injury results in lipid peroxidation and mem-
metabolism, affecting the disposition of potentially toxic
brane damage. In addition, the acetaldehyde produced by
fatty acids, have also been reported in animal experiments
the oxidation of ethanol has toxic effects, inhibiting the
(Shevchuk et al., 1991). Indeed, chronic alcohol administra-
repair of alkylated nucleoproteins (Espina et al., 1988),
tion increased omega-oxidation more effectively in male than decreasing the activity of key enzymes, and markedly reduc-
in female rats (Ma et al., 1993), explaining the potentially ing oxygen use in mitochondria damaged by long-term etha-
deleterious accumulation of nonesterified fatty acids in the nol consumption (Baraona et al., 1988; Lieber et al., 1989).
liver of the alcohol-fed female rats (Shevchuk et al., 1991). Impaired oxygen use was also confirmed in baboons in vivo
This was also associated with a much smaller ethanol-induced (Lieber et al., 1989). The impaired oxidation capacity of the
increase in cytosolic L-FABPc and microsomal esterification mitochondria may, in turn, interfere with the oxidation of
in female than in male rats, whereas the inhibition of mito- acetaldehyde (Hasumura et al., 1976), leading to a vicious
chondrial beta-oxidation was similar in both sexes. Related circle of progressive acetaldehyde accumulation and greater
findings have been observed in human beings (Ma et al., mitochondrial injury. Moreover, acetaldehyde promotes
1999) and contribute to the greater vulnerability of women cell death by depleting the concentration of reduced glutathi-
to the development of alcoholic liver injury. one (GSH), inducing lipid peroxidation, and increasing
In any event, the induction of CYP2E1 has been shown the toxic effect of free radicals. By binding to the tubulin
to play a key role in the pathogenesis of alcoholic liver of microtubules, acetaldehyde blocks the secretion of
injury, including alcoholic steatohepatitis, because of the proteins. The increases in protein, lipid, water (Wondergem
oxidative stress it generates (Lieber, 1997). In addition, & Davis, 1994), and electrolytes cause hepatocytes to
CYP2E1 is invariably elevated in the liver of patients with enlarge or “balloon,” a hallmark of alcoholic liver disease.
nonalcoholic steatohepatitis (Weltman et al., 1998) because Acetaldehyde–protein adducts promote collagen production
fatty acids (which increase in obesity) and ketones (which and may also act as neoantigens, which stimulate an immune
increase in diabetes) are also substrates for CYP2E1 (vide response (Hoerner et al., 1986; Niemela et al., 1987).
14 C.S. Lieber / Alcohol 34 (2004) 9–19

Lipid peroxidation products such as 4-hydroxynonenal variety, but excess methionine was shown to have some
stimulate fibrosis, which is also increased through decreased adverse effects, as reviewed elsewhere (Lieber, 2000).
feedback inhibition of collagen synthesis because acetalde- Whereas in some patients with alcoholic liver disease circu-
hyde forms adducts with the carboxyl-terminal propeptide of lating methionine concentrations are within normal limits, in
procollagen (Ma et al., 1997). others elevated concentrations were observed. It was reported
Oxidative stress promotes inflammation, which is aggra- that the blood clearance of methionine after an oral load
vated by an increase of the proinflammatory cytokine tumor was slowed, supporting the notion of impaired metabolism.
necrosis factor-alpha (TNF-α) in the Kupffer cells. Kupf- Indeed, for most of its functions, methionine must be acti-
fer cells are a major source of cytokines. They also harbor vated to S-adenosylmethionine (SAMe) and, in cirrhotic
CYP2E1, and its increase after chronic alcohol consumption livers, Duce et al. (1988) reported a decrease in the activity of
(Koivisto et al., 1996) may act as a major stimulator. Indeed, the enzyme involved, namely SAMe synthetase, also called
in both acute and chronic liver diseases, Kupffer cells methionine adenosyltransferase (Lieber, 2001) (Fig. 1). Fur-
become activated to produce cytokines and reactive oxygen thermore, long-term ethanol consumption in non-human pri-
radicals (Batey et al., 1998). Of potential therapeutic interest mates was associated with a significant depletion of hepatic
is the observation that dilinoleoylphosphatidylcholine SAMe (Lieber et al., 1990), which may have a number of
(DLPC) decreases stellate cell activation (Poniachik et al., adverse effects because SAMe is the principal methylating
1999) and that it selectively modulates the lipopolysaccha- agent in various transmethylation reactions, which are im-
ride-induced activation of Kupffer cells by decreasing the portant for nucleic acid and protein synthesis, membrane
production of the cytotoxic TNF-α, while potentiating the fluidity, and the transport of metabolites and transmission of
release of the protective interleukin-1 beta (IL-1β) (Oneta signals across membranes. By impairing methyltransferase
et al., 1999). This dual action of DLPC on cytokines may activity, SAMe depletion exacerbates the membrane injury
provide a potent mechanism against liver injury. In addition induced by alcohol. Furthermore, SAMe plays a key role in
to the reduction in TNF-α, the enhanced release of IL-1β the synthesis of polyamines and provides a source of cysteine
could oppose the hepatotoxicity of TNF-α, either directly for GSH production (Fig. 4).
or indirectly through an IL-1β–related increase in the toler-
ance to TNF-α–mediated toxicity.
4.1.2. Therapeutic applications of S-adenosylmethionine,
It has also been shown that ethanol induces transforming
including clinical trials
growth factor-alpha (TGF-α) production in hepatocytes,
In baboons, correction of the ethanol-induced hepatic
leading to stimulation of collagen synthesis by hepatic stel-
SAMe depletion with oral SAMe administration resulted in
late cells (Kato et al., 2003). These results support the notion
a corresponding attenuation of ethanol-induced liver injury,
that TGF-α derived from ethanol-exposed hepatocytes may
as shown by a less striking GSH depletion, lesser increases
contribute to the development of hepatic fibrosis in alcoholic
liver disease. This effect was specifically inhibited by anti–
TGF-α antibodies. It is noteworthy that, experimentally,
DLPC also decreased transforming growth factor-beta 1
(TGF-β1)–induced collagen mRNA by inhibiting p38 mito-
gen-activated protein kinase in hepatic stellate cells (Cao
et al., 2002). These are activated through induction of their
CYP2E1, through acetaldehyde, and by endotoxin. That
TNF-α is decreased by DLPC, the active phosphatidylcho-
line (PC) species of polyenylphosphatidylcholine (PPC), was
also shown by Cao et al. (2002).

4. Antifibrotic therapy through correction of altered nu-


trient activation or of the deficiency in selective nutrients Fig. 4. Link between enhanced lipid peroxidation and accelerated acetalde-
hyde production as well as increased free radical generation by the induced
The nutritional approach to liver disease has been im- microsomes, with sites of possible therapeutic interventions. Metabolic
proved by recognizing the importance of not only providing blocks caused by liver disease (a, b) or folate (c), B12 (c), or B6 (d) deficiencies
a sufficient intake of nutrients but also correcting the impair- are illustrated, with corresponding depletions in S-adenosylmethionine,
ment in the activation of key nutrients by alcohol. phosphatidylcholine, and reduced glutathione (GSH). New therapeutic ap-
proaches include down-regulation of microsomal enzyme induction,
especially of cytochrome P450 2E1 (CYP2E1); decrease of free radicals
4.1. Methionine and S-adenosylmethionine with antioxidants; and replenishment of S-adenosylmethionine and of
phosphatidylcholine. ADH ⫽ Alcohol dehydrogenase. Reprinted from
4.1.1. Pathogenesis of the deficiency and its consequences Journal of Hepatology, 32, C. S. Lieber, Alcoholic liver disease: new
Correction of methionine deficiency has been proposed insights in pathogenesis lead to new treatments, 113–128, Copyright (2000),
for the treatment of liver diseases, especially the alcoholic with permission from Elsevier.
C.S. Lieber / Alcohol 34 (2004) 9–19 15

in plasma aspartate aminotransferase and glutamic dehydro- by acetaldehyde-mediated impairments of the antioxidant
genase activities, and fewer megamitochondria (Lieber et al., systems, including depletion of GSH (Fig. 4). This results,
1990). Treatment with SAMe is also beneficial in human as reviewed elsewhere (Lieber, 1999), in peroxidation of
beings, as shown in a prospective, multicenter, double-blind, lipids, including PCs. They form the backbone of the mem-
placebo-controlled trial (Frezza et al., 1990b) performed in branes, which are strikingly altered, as illustrated by the
220 inpatients with chronic liver diseases (chronic active blebbing of the plasma membranes in alcohol-fed rats
hepatitis and cirrhosis, including primary biliary cirrhosis) (Yamada et al., 1985), as well as the distortion of the mito-
in whom serum markers of cholestasis and subjective symp- chondrial membranes in baboons and human beings consum-
toms significantly improved after SAMe treatment. Oral ad- ing alcohol, even when their diets are nutritionally adequate
ministration of SAMe at 1,200 mg/day for 6 months also (Lieber, 1992). Cytochrome P450 2E1 generates several
resulted in a significant increase of hepatic GSH concentra- species of active oxygen (Fig. 3), and GSH offers one of
tions in patients with alcoholic as well as non-alcoholic the mechanisms for the scavenging of these toxic free radi-
liver disease (Vendemiale et al., 1989). Noteworthy is a 2- cals (Fig. 4). Eventually, however, GSH is depleted, and the
year randomized, placebo-controlled, double-blind, multi- oxidative stress promotes injury by inactivation of enzymes
center clinical trial of SAMe in patients with alcoholic and peroxidation of lipids. In patients with cirrhosis, hepatic
liver cirrhosis (Child class A and B) in whom SAMe improved depletion of alpha-tocopherol (Leo et al., 1993), a major
survival (29% vs. 12%, P ⫽ .025) or delayed liver trans- antioxidant, potentiates this effect.
plantation (Mato et al., 1999).

4.2. Polyenylphosphatidylcholine and other antioxidants 4.2.2. Therapeutic approaches with PPC, including clini-
cal trials
4.2.1. Pathogenesis of the deficiency and its consequences Phosphatidylcholines were replenished in the membranes
Characteristic features of alcoholic liver injury include by administration of PPC, a mixture of polyunsaturated
striking membrane alterations with associated phospholipid PCs rich in DLPC, selected for its high bioavailability.
depletion (Lieber, 1992) as well as scarring (or fibrosis). Also, PPC totally prevented the associated septal fibrosis
Fibrosis is characterized by the accumulation of excess colla- and cirrhosis in baboons fed adequate diets (Lieber et al.,
gen after chronic ethanol consumption. This is due, at least 1994b), with an associated reduction in the number of acti-
in part, to acetaldehyde, which stimulates collagen produc- vated stellate cells. These had been found to be strikingly
tion in hepatic stellate cells (Moshage et al., 1990) (vide increased in alcohol-fed baboons, contributing to the pro-
supra). Furthermore, fibrosis is related to associated meta- gression of fibrosis (Mak et al., 1984). A similar transforma-
bolic effects of ethanol. Indeed, the hepatic oxidation of tion of stellate cells was observed in patients with alcoholic
ethanol to acetaldehyde in the ADH pathway and the further liver disease (Mak & Lieber, 1988), and an attenuation of
oxidation of acetaldehyde to acetate reduce NAD to NADH, the activation of stellate cells to myofibroblast-like cells was
thereby producing striking redox changes that have an also observed in vitro (Poniachik et al., 1999). In addition,
impact on collagen metabolism. Indeed, the rise in NADH the activity of membrane-bound enzymes was normalized:
shifts pyruvate to lactate, which stimulates collagen synthe- Cytochrome oxidase, a key enzyme of the mitochondrial
sis in cultured myofibroblasts (Savolainen et al., 1984). This electron-transport chain, which requires a normal phospho-
finding seemed to indicate that ethanol-induced redox lipid milieu for optimal activity and is severely depressed
changes may play a role, which was confirmed by the obser- by chronic ethanol consumption, was restored to normal
vation that methylene blue, a scavenger of reducing equiva- by addition of PC in vitro (Arai et al., 1984) or by PPC
lents, totally inhibits the stimulatory effect of acetaldehyde supplementation in vivo (Navder et al., 1997), with improve-
on alpha1(I) procollagen and fibronectin gene expression in ment of hepatic mitochondrial respiration.
stellate cells (Casini et al., 1991). One concern was that PPC and DLPC, because of their
Some progress was made concerning possible therapeutic polyunsaturated nature, may aggravate the oxidative stress,
effects of agents that oppose the alcohol-induced oxidative but the opposite was found, both in vitro and in vivo. In
stress. Indeed, contrary to ADH, the CYP2E1 is strikingly alcohol-fed baboons, PPC not only prevented septal fibrosis
inducible by chronic ethanol consumption, which results in and cirrhosis (Lieber et al., 1994b), but it also resulted in a
an increased production of not only acetaldehyde but also protection against oxidative stress, as determined by normal-
free radicals (Lieber, 1997). Replenishment of GSH can be ization of 4-hydroxynonenal, F2-isoprostanes, and GSH
achieved by administration of precursors of cysteine (one of concentrations (Lieber et al., 1997).
the amino acids of this tripeptide), such as acetylcysteine Even in an experimental model devoid of oxidative stress,
or SAMe (Lieber, 1999; Lieber et al., 1990). Experimentally, namely fibrosis and cirrhosis induced by the injection of
CYP2E1 has also been down-regulated by PPC (Aleynik heterologous albumin in rats, PPC was nevertheless protec-
et al., 1999) and by its main component DLPC (Aleynik tive (Ma et al., 1996). This may reflect, at least in part, that
& Lieber, 2001), a potentially beneficial therapeutic approach both PPC (Li et al., 1992) and DLPC (Lieber et al., 1994b)
(vide supra). The resulting oxidative stress is exacerbated stimulate collagenase production in stellate cells, resulting
16 C.S. Lieber / Alcohol 34 (2004) 9–19

in a decrease of collagen accumulation. The activity of phos- 5. Conclusions


phatidylethanolamine methyltransferase, a key enzyme for
Much progress has been made in the understanding of the
the regeneration of hepatic PC and which is depressed in
alcoholic liver disease (Fig. 4), was also restored with PPC pathogenesis of alcoholic liver disease, resulting in improved
treatment (Lieber et al., 1994a). prevention and therapy, with promising prospects for even
more effective treatments. The most successful approaches
All these favorable effects of PPC provided a basis for
some of the reported therapeutic effects of unsaturated phos- that one can expect to evolve are those that deal with the
fundamental cellular disturbances resulting from excessive
pholipids in the treatment of liver diseases, including in
alcohol consumption. Two pathologic concepts are emerging
patients with alcoholic hepatitis (Bird et al., 1991). Further-
more, PC interfered with collagen formation from stellate as particularly useful therapeutically. Whereas it continues
to be important to replenish nutritional deficiencies, when
cells (Lieber et al., 1994b), which have a predominant role in
present, it is crucial to recognize that, because of the alcohol-
alcohol-induced fibrogenesis. Subsequently, PPC improved
liver test results in patients with hepatitis C (Niederau et al., induced disease process, some of the nutritional require-
ments change. This is exemplified by methionine: It normally
1998) and ameliorated alcoholic liver disease in a pilot study
is one of the essential amino acids for human beings, but
of patients with underlying fibrosis (Lieber et al., 2000).
This was followed by a randomized, prospective, double- it needs to be activated to SAMe, a process impaired by
the disease. Thus, SAMe, rather than methionine, is the
blind, placebo-controlled clinical trial conducted in 20
compound that must be supplemented in the presence of
Veterans Affairs Medical Centers with 789 patients (97%
male; mean age, 48.8 years) averaging 16 drinks per day significant liver disease. Indeed, SAMe was found to attenu-
ate mitochondrial lesions in baboons, replenish GSH, and
(1 drink ⫽ 14 g of alcohol) for 19 years (Lieber et al., 2003b,
2003c). Evaluation of a baseline liver biopsy sample con- significantly reduce mortality in patients with Child A or B
firmed the presence of perivenular or septal fibrosis or cirrhosis. Similarly, PPC corrects the ethanol-induced he-
patic phospholipid depletion, restores fully or partially
incomplete cirrhosis. Alcohol intake was reduced in both
groups to approximately 2 1/2 drinks per day as a result phosphatidylethanolamine methyltransferase activity, and
of the “Brief Intervention” approach (Lieber et al., 2003b, opposes oxidative stress. It also deactivates hepatic stellate
cells, whereas its DLPC increases collagenase activity, result-
2003c). Accordingly, there was no further progression of
the fibrosis and, therefore, no way to test whether PPC could ing in prevention of ethanol-induced septal fibrosis and
cirrhosis in the baboon. Furthermore, CYP2E1, when ex-
oppose such a progression, except in a subgroup who were
cessively induced, becomes harmful and should be down-
still consuming 6 or more drinks a day and showed benefi-
cial effects against fibrosis. Ascites, an important secondary regulated. PPC is one of the substances with anti-CYP2E1
properties that is now emerging (Aleynik & Lieber, 2001).
clinical measure of liver disease, was also observed less
A quarter of all patients with alcoholic liver disease are
frequently during monitoring of PPC-treated patients. Fur-
thermore, improvement in concentrations of transaminases seropositive for hepatitis C virus, with an even higher inci-
dence in some urban areas. In addition to antiviral medica-
and bilirubin favoring PPC was seen at some time points in
tions, agents that oppose oxidative stress and fibrosis are
the subgroups of drinkers who were seropositive for hepatitis
C virus. being tested for hepatitis C treatment because these two
processes contribute much to the pathologic changes and
mortality associated with the virus.
4.2.3. Silymarin and other antioxidants
Silymarin was found to be effective against various liver Thus, effective prevention and therapy against steatosis
and its progression to more severe injury can be achieved
injuries in rodents. In patients with alcoholic liver disease,
results of some randomized controlled trials with silymarin by a multifactorial approach: control of alcohol consump-
showed beneficial effects such as improved survival (Ferenci tion, avoidance of obesity and excess dietary long-chain fatty
acids, or their replacement with medium-chain triglycerides,
et al., 1989). However, findings of other studies did not
verify such an effect (Parés et al., 1998). To clarify this issue, and replenishment of SAMe and the PCs.
silymarin was studied under controlled conditions in non-
human primates, and it was found to oppose the alcohol- Acknowledgments
induced oxidative stress and to retard the development of
Original studies were supported, in part, by NIH grants
alcohol-induced hepatic fibrosis (Lieber et al., 2003a). The
AA11115 and AT001583; the Department of Veterans Af-
negative outcome observed in some clinical trials possibly
fairs; and the Kingsbridge and C.D. Smithers Research Foun-
reflects poor compliance. Thus, in view of the innocuity of
dations. I am grateful to Ms. Y. Rodriguez for the skillful
silymarin, it might be justified to verify its effect in additional
typing of this document.
clinical studies. Theoretically, other antioxidants such as
alpha-tocopherol might be useful. Indeed, in patients with
cirrhosis, diminished hepatic vitamin E concentrations have References
been observed (Leo et al., 1993). However, therapeutic Adas, F., Berthou, F., Picart, D., Lozac’h, P., Beaugé, F., & Amet, Y. (1998).
results were disappointing (de la Maza et al., 1995). Involvement of cytochrome P450 2E1 in the (ω–1)-hydroxylation
C.S. Lieber / Alcohol 34 (2004) 9–19 17

of oleic acid in human and rat liver microsomes. J Lipid Res 39, Eriksson, C. J. P., Fukunaga, T., Sarkola, T., Lindholm, H., & Ahola,
1210–1219. L. (1996). Estrogen-related acetaldehyde elevation in women during
Aleynik, M. K., Leo, M. A., Aleynik, S. I., & Lieber, C. S. (1999). Polyenyl- alcohol intoxication. Alcohol Clin Exp Res 20, 1192–1195.
phosphatidylcholine opposes the increase of cytochrome P-4502E1 by Espina, N., Lima, V., Lieber, C. S., & Garro, A. J. (1988). In vitro and in
ethanol and corrects its iron-induced decrease. Alcohol Clin Exp Res vivo inhibitory effect of ethanol and acetaldehyde on O6-methylguanine
23, 96–100. transferase. Carcinogenesis 9, 761–766.
Aleynik, M. K., & Lieber, C. S. (2001). Dilinoleoylphosphatidylcholine Ferenci, P., Dragosics, B., Dittrich, H., Frank, H., Benda, L., Lochs, H.,
decreases ethanol-induced cytochrome P4502E1. Biochem Biophys Res Meryn, S., Base, W., & Schneider, B. (1989). Randomized controlled
Commun 288, 1047–1051. trial of silymarin treatment in patients with cirrhosis of the liver. J
Amet, Y., Berthou, F., Goasduff, T., Salaun, J. P., Le Breton, L., & Menez, Hepatol 9, 105–113.
J. F. (1994). Evidence that cytochrome P450 2E1 is involved in the Frezza, M., di Padova, C., Pozzato, G., Terpin, M., Baraona, E., & Lieber,
(ω-1)-hydroxylation of lauric acid in rat liver microsomes. Biochem C. S. (1990a). High blood alcohol levels in women. The role of decreased
Biophys Res Commun 203, 1168–1174. gastric alcohol dehydrogenase activity and first-pass metabolism. N
Angulo, P., & Lindor, K. D. (2001). Insulin resistance and mitochondrial Engl J Med 322, 95–99.
abnormalities in NASH: a cool look into a burning issue. Gastroenter- Frezza, M., Surrenti, C., Manzillo, G., Fiaccadori, F., Bortolini, M., & Di
ology 120, 1281–1285 (Editorial). Padova, C. (1990b). Oral S-adenosylmethionine in the symptomatic
Arai, M., Gordon, E. R., & Lieber, C. S. (1984). Decreased cytochrome treatment of intrahepatic cholestasis. A double-blind, placebo-controlled
oxidase activity in hepatic mitochondria after chronic ethanol consump- study. Gastroenterology 99, 211–215.
tion and the possible role of decreased cytochrome aa3 content and Galli, A., Price, D., & Crabb, D. (1999). High-level expression of rat class
changes in phospholipids. Biochim Biophys Acta 797, 320–327. I alcohol dehydrogenase is sufficient for ethanol-induced fat accumula-
Baraona, E., Matsumura, T., Hernandez, R., Kubota, S., Soong, J., Kawano, tion in transduced HeLa cells. Hepatology 29, 1164–1170.
S., Kato, S., Inatomi, N., Sato, S., & Lieber, C.S. (1988). Chronic Hasumura, Y., Teschke, R., & Lieber, C. S. (1976). Characteristics of
alcohol intake impairs liver oxygenation by decreasing O2 utilization. acetaldehyde oxidation in rat liver mitochondria. J Biol Chem 251,
In K. Kuriyama, A. Takada, & H. Ishii (Eds.), Biomedical and Social 4908–4913.
Aspects of Alcohol and Alcoholism: Proceedings of the Fourth Congress Hoerner, M., Behrens, U. J., Worner, T., & Lieber, C. S. (1986). Humoral
of the International Society for Biomedical Research on Alcoholism immune response to acetaldehyde adducts in alcoholic patients. Res
(pp. 387–390). Amsterdam, The Netherlands: Elsevier Science Publish- Commun Chem Pathol Pharmacol 54, 3–12.
ers BV. Iseri, O. A., Gottlieb, L. S., & Lieber, C. S. (1964). The ultrastructure of
Batey, R., Cao, Q., Madsen, G., Pang, G., Russell, A., & Clancy, R. ethanol induced fatty liver. Fed Proc 23, 579A (Abstract).
(1998). Decreased tumor necrosis factor-α and interleukin-1α production Iseri, O. A., Lieber, C. S., & Gottlieb, L. S. (1966). The ultrastructure of
from intrahepatic mononuclear cells in chronic ethanol consumption and fatty liver induced by prolonged ethanol ingestion. Am J Pathol 48,
upregulation by endotoxin. Alcohol Clin Exp Res 22, 150–156. 535–555.
Best, C. H., Hartroft, W. S., Lucas, C. C., & Ridout, J. H. (1949). Liver Ishii, H., Joly, J.-G., & Lieber, C. S. (1973a). Effect of ethanol on the
damage produced by feeding alcohol or sugar and its prevention by amount and enzyme activities of hepatic rough and smooth microsomal
choline. Br Med J 2, 1001–1006. membranes. Biochim Biophys Acta 291, 411–420.
Bird, G. L. A., Panos, M. Z., Polson, R., Johnson, R., Portman, B., Neu- Ishii, H., Joly, J.-G., & Lieber, C. S. (1973b). Increase of microsomal
berger, J., & Williams, R. (1991). Activity of polyunsaturated phosphati- glucose-6-phosphatase activity after chronic ethanol administration.
dylcholine in HbsAg negative chronic active hepatitis and acute Metabolism 22, 799–806.
alcoholic hepatitis. Z Gastroenterol 29, 21–24. Isselbacher, K. J., & Carter, E. A. (1970). Ethanol oxidation by liver
Boleda, M. D., Saubi, N., Farrés, J., & Parés, X. (1993). Physiological microsomes: evidence against a separate and distinct enzyme system.
substrates for rat alcohol dehydrogenase classes: aldehydes of lipid
Biochem Biophys Res Commun 39, 530–537.
peroxidation, ω-hydroxyfatty acids, and retinoids. Arch Biochem Bio-
James, O. F. W., & Day, C. P. (1998). Non-alcoholic steatohepatitis (NASH):
phys 307, 85–90.
a disease of emerging identity and importance. J Hepatol 29, 495–501.
Burnett, D. A., Lysenko, N., Manning, J. A., & Ockner, R. K. (1979).
Johansson, I., Eliasson, E., Norsten, C., & Ingelman-Sundberg, M. (1986).
Utilization of long chain fatty acids by rat liver: studies of the role of
Hydroxylation of acetone by ethanol- and acetone-inducible cytochrome
fatty acid binding protein. Gastroenterology 77, 241–249.
P-450 in liver microsomes and reconstituted membranes. FEBS Lett
Cao, Q., Mak, K. M., & Lieber, C. S. (2002). DLPC decreases TGF-
196, 59–64.
β1-induced collagen mRNA by inhibiting p38 MAPK in hepatic stellate
Jones, B. M., & Jones, M. K. (1976). Male and female intoxication levels
cells. Am J Physiol Gastrointest Liver Physiol 283, G1051–G1061.
for three alcohol doses or do women really get higher than men?
Casazza, J. P., Felver, M. E., & Veech, R. L. (1984). The metabolism of
Alcohol Technical Report 5, 544–583.
acetone in rat. J Biol Chem 259, 231–236.
Kaikaus, R. M., Chan, W. K., Ortiz de Montellano, P. R., & Bass, N. M.
Casazza, J. P., & Veech, R. L. (1985). The production of 1,2-propanediol
(1993). Mechanisms of regulation of liver fatty acid–binding protein.
in ethanol treated rats. Biochem Biophys Res Commun 129, 426–430.
Mol Cell Biochem 123, 93–100.
Casini, A., Cunningham, M., Rojkind, M., & Lieber, C. S. (1991). Acetalde-
hyde increases procollagen type I and fibronectin gene transcription Kato, J., Sato, Y., Inui, N., Nakano, Y., Takimoto, R., Takada, K., Kobune,
in cultured rat fat-storing cells through a protein synthesis-dependent M., Kuroiwa, G., Miyake, S., Kohgo, Y., & Niitsu, Y. (2003). Ethanol
mechanism. Hepatology 13, 758–765. induces transforming growth factor-α expression in hepatocytes, leading
Chedid, A., Mendenhall, C. L., Gartside, P., French, S. W., Chen, T., Rabin, to stimulation of collagen synthesis by hepatic stellate cells. Alcohol
L., and the VA Cooperative Study Group. (1991). Prognostic factors Clin Exp Res 27(8 Suppl), 58S–63S.
in alcoholic liver disease. Am J Gastroenterol 86, 210–216. Khani, S. C., Porter, T. D., & Coon, M. J. (1988). Isolation and partial
Conney, A. H. (1967). Pharmacological implications of microsomal enzyme characterization of the gene for cytochrome P-450 3a (P-450ALC) and
induction. Pharmacol Rev 19, 317–366. a second closely related gene. Biochem Biophys Res Commun 150,
de la Maza, M. P., Petermann, M., Bunout, D., & Hirsch, S. (1995). Effects 10–17.
of long-term vitamin E supplementation in alcoholic cirrhotics. J Am Koivisto, T., Mishin, V. M., Mak, K. M., Cohen, P. A., & Lieber, C. S.
Coll Nutr 14, 192–196. (1996). Induction of cytochrome P-4502E1 by ethanol in rat Kupffer
Duce, A. M., Ortiz, P., Cabrero, C., & Mato, J. M. (1988). S-adenosyl-L- cells. Alcohol Clin Exp Res 20, 207–212.
methionine synthetase and phospholipid methyltransferase are inhibited Koop, D. R. (1992). Oxidative and reductive metabolism by cytochrome
in human cirrhosis. Hepatology 8, 65–68. P450 2E1. FASEB J 6, 724–730.
18 C.S. Lieber / Alcohol 34 (2004) 9–19

Koop, D. R., & Casazza, J. P. (1985). Identification of ethanol-inducible Lieber, C. S., Leo, M. A., Aleynik, S. I., Lowe, N., Aleynik, M. K., &
P-450 isozyme 3a as the acetone and acetol monooxygenase of rabbit Paronetto, F. (2000). Increasing circulating level of dilinoleoylphospha-
microsomes. J Biol Chem 260, 13607–13612. tidylcholine is associated with protection against alcohol induced oxi-
Koop, D. R., & Tierney, D. J. (1990). Multiple mechanisms in the regulation dative stress and liver fibrosis in man. Hepatology 32, 386A (Abstract).
of ethanol-inducible cytochrome P450IIE1. Bioessays 12, 429–435. Lieber, C. S., Leo, M. A., Cao, Q., Ren, C., & DeCarli, L. M. (2003a).
Laethem, R. M., Balazy, M., Falck, J. R., Laethem, C. L., & Koop, D. R. Silymarin retards the progression of alcohol-induced hepatic fibrosis
(1993). Formation of 19(S)-, 19(R)-, and 18(R)-hydroxyeicosatetraenoic in baboons. J Clin Gastroenterol 37, 336–339.
acids by alcohol-inducible cytochrome P450 2E1. J Biol Chem 268, Lieber, C. S., Robins, S. J., & Leo, M. A. (1994a). Hepatic phosphatidyletha-
12912–12918. nolamine methytransferase activity is decreased by ethanol and in-
Lane, B. P., & Lieber, C. S. (1966). Ultrastructural alterations in human creased by phosphatidylcholine. Alcohol Clin Exp Res 18, 592–595.
hepatocytes following ingestion of ethanol with adequate diets. Am J Lieber, C. S., Robins, S. J., Li, J., DeCarli, L. M., Mak, K. M., Fasulo, J. M., &
Pathol 49, 593–603. Leo, M. A. (1994b). Phosphatidylcholine protects against fibrosis and
Lane, B. P., & Lieber, C. S. (1967). Effects of butylated hydroxytoluene cirrhosis in the baboon. Gastroenterology 106, 152–159.
on the ultrastructure of rat hepatocytes. Lab Invest 16, 342–348. Lieber, C. S., & Schmid, R. (1961). The effect of ethanol on fatty acid
Leo, M. A., Rosman, A. S., & Lieber, C. S. (1993). Differential depletion metabolism: stimulation of hepatic fatty acid synthesis in vitro. J Clin
of carotenoids and tocopherol in liver disease. Hepatology 17, 977–986. Invest 40, 394–399.
Li, J., Kim, C. I., Leo, M. A., Mak, K. M., Rojkind, M., & Lieber, C. S. Lieber, C. S., Weiss, D. G., Groszmann, R., Paronetto, F., & Schenker, S.,
for the Veterans Affairs Cooperative Study 391 Group. (2003b). I.
(1992). Polyunsaturated lecithin prevents acetaldehyde-mediated he-
Veterans Affairs Cooperative Study of polyenylphosphatidylcholine
patic collagen accumulation by stimulating collagenase activity in cul-
in alcoholic liver disease: effects on drinking behavior by nurse/physician
tured lipocytes. Hepatology 15, 373–381.
teams. Alcohol Clin Exp Res 27, 1757–1764.
Lieber, C. S. (1992). Medical and Nutritional Complications of Alcoholism:
Lieber, C. S., Weiss, D. G., Groszmann, R., Paronetto, F., & Schenker, S.,
Mechanisms and Management. New York: Plenum Press.
for the Veterans Affairs Cooperative Study 391 Group. (2003c). II.
Lieber, C. S. (1997). Cytochrome P-4502E1: its physiological and patholog-
Veterans Affairs Cooperative Study of polyenylphosphatidylcholine in
ical role. Physiol Rev 77, 517–544. alcoholic liver disease. Alcohol Clin Exp Res 27, 1765–1772.
Lieber, C. S. (1999). Microsomal ethanol-oxidizing system (MEOS): the Ma, X., Baraona, E., Goozner, B. G., & Lieber, C. S. (1999). Gender
first 30 years (1968–1998)—a review. Alcohol Clin Exp Res 23, differences in medium-chain dicarboxylic aciduria in alcoholic men and
991–1007. women. Am J Med 106, 70–75.
Lieber, C. S. (2000). Alcoholic liver disease: new insights in pathogenesis Ma, X., Baraona, E., & Lieber, C. S. (1993). Alcohol consumption enhances
lead to new treatments. J Hepatol 32(suppl 1), 113–128. fatty acid ω-oxidation, with a greater increase in male than in female
Lieber, C. S. (2001). Alcoholic liver injury: pathogenesis and therapy in rats. Hepatology 18, 1247–1253.
2001. Pathol Biol (Paris) 49, 738–752. Ma, X., Svegliati-Baroni, G., Poniachik, J., Baraona, E., & Lieber, C. S.
Lieber, C. S., Baraona, E., Hernández-Muñoz, R., Kubota, S., Sato, N., (1997). Collagen synthesis by liver stellate cells is released from its
Kawano, S., Matsumura, T., & Inatomi, N. (1989). Impaired oxygen uti- normal feedback regulation by acetaldehyde-induced modification of
lization: a new mechanism for the hepatotoxicity of ethanol in sub- the carboxyl-terminal propeptide of procollagen. Alcohol Clin Exp Res
human primates. J Clin Invest 83, 1682–1690. 21, 1204–1211.
Lieber, C. S., Casini, A., DeCarli, L. M., Kim, C. I., Lowe, N., Sasaki, Ma, X., Zhao, J., & Lieber, C. S. (1996). Polyenylphosphatidylcholine
R., & Leo, M. A. (1990). S-adenosyl-L-methionine attenuates alcohol- attenuates non-alcoholic hepatic fibrosis and accelerates its regression.
induced liver injury in the baboon. Hepatology 11, 165–172. J Hepatol 24, 604–613.
Lieber, C. S., & DeCarli, L. M. (1968). Ethanol oxidation by hepatic Mak, K. M., Leo, M. A., & Lieber, C. S. (1984). Alcoholic liver injury in
microsomes: adaptive increase after ethanol feeding. Science 162, baboons: transformation of lipocytes to transitional cells. Gastroenterol-
917–918. ogy 87, 188–200.
Lieber, C. S., & DeCarli, L. M. (1970). Quantitative relationship between Mak, K. M., & Lieber, C. S. (1988). Lipocytes and transitional cells in
amount of dietary fat and severity of the alcoholic fatty liver. Am J alcoholic liver disease: a morphometric study. Hepatology 8, 1027–
Clin Nutr 23, 474–478. 1033.
Lieber, C. S., & DeCarli, L. M. (1974a). An experimental model of alcohol Mato, J. M., Cámara, J., Fernández de Paz, J., Caballerı́a, L., Coll, S.,
feeding and liver injury in the baboon. J Med Primatol 3, 153–163. Caballero, A., Garcı́a-Buey, L., Beltrán, J., Benita, V., Caballerı́a, J., Solà,
Lieber, C. S., & DeCarli, L. M. (1974b). Oxidation of ethanol by hepatic R., Moreno-Otero, R., Barrao, F., Martı́n-Duce, A., Correa, J. A., Parés,
microsomes of acatalasemic mice. Biochem Biophys Res Commun 60, A., Barrao, E., Garcı́a-Magaz, I., Puerta, J. L., Moreno, J., Boissard,
1187–1192. G., Ortiz, P., & Rodés, J. (1999). S-Adenosylmethionine in alcoholic
Lieber, C. S., DeCarli, L. M., & Schmid, R. (1959). Effects of ethanol on liver cirrhosis: a randomized, placebo-controlled, double-blind, multi-
center clinical trial. J Hepatology 30, 1081–1089.
fatty acid metabolism in liver slices. Biochem Biophys Res Commun
Meldolesi, J. (1967). On the significance of the hypertrophy of the smooth
1, 302–306.
endoplasmic reticulum in liver cells after administration of drugs.
Lieber, C. S., Jones, D. P., & DeCarli, L. M. (1965). Effects of prolonged
Biochem Pharmacol 16, 125–131.
ethanol intake: production of fatty liver despite adequate diets. J Clin
Mishra, L., Sharma, S., Potter, J. J., & Mezey, E. (1989). More rapid
Invest 44, 1009–1021.
elimination of alcohol in women as compared to their male siblings.
Lieber, C. S., Jones, D. P., Mendelson, J., & DeCarli, L. M. (1963). Fatty Alcohol Clin Exp Res 13, 752–754.
liver, hyperlipemia and hyperuricemia produced by prolonged alcohol Miwa, G. T., Levin, W., Thomas, P. E., & Lu, A. Y. H. (1978). The direct
consumption, despite adequate dietary intake. Trans Assoc Am Physi- oxidation of ethanol by a catalase- and alcohol dehydrogenase–free
cians 76, 289–300. reconstituted system containing cytochrome P-4501. Arch Biochem
Lieber, C. S., Lefèvre, A., Spritz, N., Feinman, L., & DeCarli, L. M. Biophys 187, 464–475.
(1967). Difference in hepatic metabolism of long- and medium-chain Moshage, H., Casini, A., & Lieber, C. S. (1990). Acetaldehyde selectively
fatty acids: the role of fatty acid chain length in the production of the stimulates collagen production in cultured rat liver fat-storing cells but
alcoholic fatty liver. J Clin Invest 46, 1451–1460. not in hepatocytes. Hepatology 12(3 Pt 1), 511–518.
Lieber, C. S., Leo, M. A., Aleynik, S. I., Aleynik, M. K., & DeCarli, Navder, K. P., Baraona, E., & Lieber, C. S. (1997). Polyenylphosphatidyl-
L. M. (1997). Polyenylphosphatidylcholine decreases alcohol-induced choline attenuates alcohol-induced fatty liver and hyperlipemia in rats.
oxidative stress in the baboon. Alcohol Clin Exp Res 21, 375–379. J Nutr 127, 1800–1806.
C.S. Lieber / Alcohol 34 (2004) 9–19 19

Nelson, D. R., Kamataki, T., Waxman, D. J., Guengerich, F. P., Estabrook, Tephly, T. R., Tinelli, F., & Watkins, W. D. (1969). Alcohol metabolism:
R. W., Feyereisen, R., Gonzalez, F. J., Coon, M. J., Gunsalus, I. C., role of microsomal oxidation in vivo. Science 166, 627–628.
Gotoh, O., Okuda, K., & Nebert, D. W. (1993). The P450 superfamily: Teschke, R., Hasumura, Y., Joly, J.-G., & Lieber, C. S. (1972). Microsomal
update on new sequences, gene mapping, accession numbers, early trivial ethanol-oxidizing system (MEOS): purification and properties of a rat
names of enzymes, and nomenclature. DNA Cell Biol 12, 1–51. liver system free of catalase and alcohol dehydrogenase. Biochem Bio-
Niederau, C., Strohmeyer, G., Heintges, T., Peter, K., & Gopfert, E., for phys Res Commun 49, 1187–1193.
Leich Study Group. (1998). Polyunsaturated phosphatidyl-choline and Teschke, R., Hasumura, Y., & Lieber, C. S. (1975). Hepatic microsomal
interferon alpha for treatment of chronic hepatitis B and C: a multi- alcohol-oxidizing system in normal and acatalasemic mice: its dissocia-
center, randomized, double-blind, placebo-controlled trial. Hepatogas- tion from the peroxidatic activity of catalase-H2O2. Mol Pharmacol 11,
troenterology 45, 797–804. 841–849.
Niemela, O., Klajner, F., Orrego, H., Vidins, E., Blendis, L., & Israel, Y. Teschke, R., Matsuzaki, S., Ohnishi, K., DeCarli, L. M., & Lieber, C. S.
(1987). Antibodies against acetaldehyde-modified protein epitopes in (1977). Microsomal ethanol oxidizing system (MEOS): current status
human alcoholics. Hepatology 7, 1210–1214. of its characterization and its role. Alcohol Clin Exp Res 1, 7–15.
Ohnishi, K., & Lieber, C. S. (1977). Reconstitution of the microsomal Thurman, R. G., & Brentzel, H. J. (1977). The role of alcohol dehydrogenase
ethanol-oxidizing system: qualitative and quantitative changes of cyto- in microsomal ethanol oxidation and the adaptive increase in ethanol
chrome P-450 after chronic ethanol consumption. J Biol Chem 252, metabolism due to chronic treatment with ethanol. Alcohol Clin Exp
7124–7131. Res 1, 33–38.
Oneta, C. M., Mak, K. M., & Lieber, C. S. (1999). Dilinoleoylphosphatidyl- Thurman, R. G., Ley, H. G., & Scholz, R. (1972). Hepatic microsomal
choline selectively modulates lipopolysaccharide-induced Kupffer cell ethanol oxidation. Hydrogen peroxide formation and the role of catalase.
activation. J Lab Clin Med 134, 466–470. Eur J Biochem 25, 420–430.
Parés, A., Planas, R., Torres, M., Caballerı́a, J., Viver, J. M., Acero, D., Panés, Tsutsumi, M., Lasker, J. M., Shimizu, M., Rosman, A. S., & Lieber, C. S.
J., Rigau, J., Santos, J., & Rodés, J. (1998). Effects of silymarin in (1989). The intralobular distribution of ethanol-inducible P450IIE1 in
alcoholic patients with cirrhosis of the liver: results of a controlled, rat and human liver. Hepatology 10, 437–446.
double-blind, randomized and multicenter trial. J Hepatol 28, 615–621. Umeno, M., McBride, O. W., Yang, C. S., Gelboin, H. V., & Gonzalez, F. J.
Poniachik, J., Baraona, E., Zhao, J., & Lieber, C. S. (1999). Dilinoleoylphos-
(1988a). Human ethanol-inducible P450IIE1: complete gene sequence,
phatidylcholine decreases hepatic stellate cell activation. J Lab Clin
promoter characterization, chromosome mapping, and cDNA-directed
Med 133, 342–348.
expression. Biochemistry 27, 9006–9013.
Raucy, J. L., Lasker, J. M., Kraner, J. C., Salazar, D. E., Lieber, C. S., &
Umeno, M., Song, B. J., Kozak, C., Gelboin, H. V., & Gonzalez, F. J.
Corcoran, G. B. (1991). Induction of cytochrome P450IIE1 in the obese
(1988b). The rat P450IIE1 gene: complete intron and exon sequence,
overfed rat. Mol Pharmacol 39, 275–280.
chromosome mapping, and correlation of developmental expression
Reichard, G. A. Jr., Skutches, C. L., Hoeldtke, R. D., & Owen, O. E.
with specific 5′ cytosine demethylation. J Biol Chem 263, 4956–4962.
(1986). Acetone metabolism in humans during diabetic ketoacidosis.
Vatsis, K. P., & Schulman, M. P. (1973). Absence of ethanol metabolism
Diabetes 35, 668–674.
in “acatalatic” hepatic microsomes that oxidize drugs. Biochem Biophys
Roach, M. K., Resse, W. N. Jr., & Creaven, P. J. (1969). Ethanol oxidation
in the microsomal fraction of rat liver. Biochem Biophys Res Commun Res Commun 52, 588–594.
36, 596–602. Vatsis, K. P., & Schulman, M. P. (1974). An unidentified constituent of
Salmela, K. S., Kessova, I. G., Tsyrlov, I. B., & Lieber, C. S. (1998). ethanol oxidation in hepatic microsomes. Fed Proc 33, 554A (Abstract).
Respective roles of human cytochrome P-4502E1, 1A2, and 3A4 in Vendemiale, G., Altomare, E., Trizio, T., Le Grazie, C., Di Padova, C.,
the hepatic microsomal ethanol oxidizing system. Alcohol Clin Exp Res Salerno, M. T., Carrieri, V., & Albano, O. (1989). Effects of oral
22, 2125–2132. S-adenosyl-L-methionine on hepatic glutathione in patients with liver
Sanyal, A. J., Campbell-Sargent, C., Mirshahi, F., Rizzo, W. B., Contos, disease. Scand J Gastroenterol 24, 407–415.
M. J., Sterling, R. K., Luketic, V. A., Shiffman, M. L., & Clore, J. N. Weltman, M. D., Farrell, G. C., Hall, P., lngelman-Sundberg, M., & Liddle,
(2001). Nonalcoholic steatohepatitis: association of insulin resistance C. (1998). Hepatic cytochrome P450 2E1 is increased in patients with
and mitochondrial abnormalities. Gastroenterology 120, 1183–1192. nonalcoholic steatohepatitis. Hepatology 27, 128–133.
Savolainen, E. R., Leo, M. A., Timpl, R., & Lieber, C. S. (1984). Acetalde- Wondergem, R., & Davis, J. (1994). Ethanol increases hepatocyte water
hyde and lactate stimulate collagen synthesis of cultured baboon liver volume. Alcohol Clin Exp Res 18, 1230–1236.
myofibroblasts. Gastroenterology 87, 777–787. Yamada, S., Mak, K. M., & Lieber, C. S. (1985). Chronic ethanol consump-
Shevchuk, O., Baraona, E., Ma, X. L., Pignon, J. P., & Lieber, C. S. tion alters rat liver plasma membranes and potentiates release of alkaline
(1991). Gender differences in the response of hepatic fatty acids and phosphatase. Gastroenterology 88, 1799–1806.
cytosolic fatty acid–binding capacity to alcohol consumption in rats. Yang, C. S., Yoo, J. S., Ishizaki, H., & Hong, J. Y. (1990). Cytochrome
Proc Soc Exp Biol Med 198, 584–590. P450IIE1: roles in nitrosamine metabolism and mechanisms of regula-
Takahashi, T., Lasker, J. M., Rosman, A. S., & Lieber, C. S. (1993). Induction tion. Drug Metab Rev 22, 147–159.
of cytochrome P-4502E1 in the human liver by ethanol is caused by Ziegler, D. M. (1972). Discussion. In R. W. Estabrook, R. J. Gillette, &
a corresponding increase in encoding messenger RNA. Hepatology 17, K. C. Lieberman (Eds.), Microsomes and Drug Oxidations (p. 458).
236–245. Baltimore: Williams & Wilkins.

You might also like