Gastro-Liver Phisiology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Am J Physiol Gastrointest Liver Physiol 287: G1–G6, 2004;

10.1152/ajpgi.00056.2004. Theme

Recent Advances in Alcoholic Liver Disease


II. Minireview: molecular mechanisms of alcoholic fatty liver
Min You and David W. Crabb
Departments of Medicine, Indiana University School of Medicine and Richard
Roudebush Veteran’s Affairs Medical Center, Indianapolis, Indiana 46202

You, Min, and David W. Crabb. Recent Advances in Alcoholic of peroxisomal ␤-oxidation enzymes; however, the receptor is
Liver Disease. II. Minireview: molecular mechanisms of alcoholic required for induction of this system by peroxisome prolifera-
fatty liver. Am J Physiol Gastrointest Liver Physiol 287: G1–G6, tors (1). PPAR␣ knockout animals develop fatty liver and
2004; 10.1152/ajpgi.00056.2004.—Alcohol has long been thought to obesity as they age (5) or are fed a high-fat diet (10). They are
cause fatty liver by way of altered NADH/NAD⫹ redox potential in
particularly sensitive to the development of steatohepatitis
the liver, which, in turn, inhibits fatty acid oxidation and the activity
of tricarboxylic acid cycle reactions. More recent studies indicate that when fed a methionine- and choline-deficient diet (8). This
additional effects of ethanol both impair fat oxidation and stimulate may relate to the inability of the liver to export fat in the form
lipogenesis. Ethanol interferes with DNA binding and transcription- of lipoproteins when methionine and choline are deficient.
activating properties of peroxisome proliferator-activated receptor-␣ Fatty acid levels are markedly increased in the liver after
(PPAR␣), as demonstrated with cultured cells and in ethanol-fed alcohol consumption; therefore, one would predict that the
mice. Treatment of ethanol-fed mice with a PPAR␣ agonist can PPAR␣-controlled battery of fat-metabolizing enzymes should
reverse fatty liver even in the face of continued ethanol consumption. be induced by alcohol consumption. Whereas a subset of
Ethanol also activated sterol regulatory element binding protein 1, PPAR␣-responsive genes has been reported to be induced by
inducing a battery of lipogenic enzymes. These effects may be due in ethanol, for example, cytochrome P-450 4A1 (lauryl ␻-hy-
part to inhibition of AMP-dependent protein kinase, reduction in droxylase) and liver fatty acid binding protein, many others did
plasma adiponectin, or increased levels of TNF-␣ in the liver. The
not change or even decreased; for example, medium-chain
understanding of these ethanol effects provides new therapeutic tar-
gets to reverse alcoholic fatty liver. acyl-CoA dehydrogenase activity and mRNA levels were de-
creased by ethanol feeding. The physiological result of the
triglyceride; lipid; transcription factor; ethanol increased generation of dicarboxylic fatty acids by enhanced
lauryl ␻-hydroxylase activity and the failure of ethanol to
induce acyl-CoA oxidase, the first step in peroxisomal ␤-oxi-
THE UNDERSTANDING OF THE CONTROL of lipid metabolism in the dation, is increased excretion of urinary dicarboxylic fatty
liver and other tissues has undergone a revolution in the past acids in alcohol-fed rats and in alcoholic men. Thus it would
decade. The control of levels of the enzymes involved in fatty appear that the failure of full induction of fatty acid-metabo-
acid oxidation and synthesis are predominantly controlled by lizing genes in alcohol-fed animals might contribute to the
two regulatory molecules, peroxisome proliferator-activated development of fatty liver.
receptor-␣ (PPAR␣) and sterol regulatory element binding The rate of synthesis of fat is also increased by ethanol. This
protein 1 (SREBP-1), respectively (Fig. 1). PPAR␣ is a mem- is accompanied by increased levels of a number of lipogenic
ber of the nuclear hormone receptor superfamily, which, when enzymes, which are now known to be regulated by the tran-
dimerized with retinoid X receptors (RXR), controls transcrip- scription factor SREBP. SREBP belong to a family of tran-
tion of a set of genes containing peroxisome proliferator scription factors containing basic helix-loop-helix-leucine zip-
response elements (PPREs), which are involved in free fatty per motifs and regulate enzymes responsible for cholesterol,
acid (FFA) transport and oxidation. These include membrane fatty acid, and triglyceride synthesis in liver and other tissues.
transporters such as carnitine palmitoyl-transferase I (CPT I), Three SREBP isoforms, designated SREBP-1a, SREBP-1c,
apolipoprotein genes, and several components of the mitochon- and SREBP-2, have been identified. SREBP-1a is mainly
drial and peroxisomal fatty acid oxidation pathways. In addi- expressed in cultured cells, providing both cholesterol and fatty
tion, an enzyme that is critical to the control of malonyl-CoA acids that are required for cell membrane synthesis. In liver,
levels, malonyl-CoA decarboxylase (MCD), was recently SREBP-1c is involved in fatty acid synthesis, whereas
shown to be regulated by PPAR␣ (12). The transcriptional SREBP-2 plays a major role in the regulation of cholesterol
activity of PPAR␣ is activated on binding of FFA as well as a synthesis. SREBPs are synthesized as 125-kDa precursors and
number of drugs such as nonsteroidal anti-inflammatory drugs are attached to the nuclear envelope and endoplasmic reticulum.
and fibrates. This provides a feedback loop that increases the On activation, the NH2-terminal domain of each SREBP (68 kDa,
capacity of the liver to dispose of fatty acids when the intra- referred as mature protein) is released proteolytically from the
cellular concentration of the fatty acids rises. Fasted PPAR␣- membrane into the nucleus, where it binds to and activates
null mice have severe impairment in their ability to oxidize promoters containing sterol response elements (SREs). The role of
FFA in the liver, resulting in hypoglycemia, hypothermia, SREBP-1 in fatty liver development has been firmly established
hypoketonemia, elevated plasma levels of FFAs, and fatty liver by animal models, including transgenic and knockout mice.
(1). PPAR␣ is not required for expression of constitutive levels Transgenic mice overexpressing SREBP-1a or -1c have massive

Address for reprint requests and other correspondence: D. W. Crabb, The costs of publication of this article were defrayed in part by the payment
Emerson Hall Rm. 317, 545 Barnhill Dr., Indianapolis IN 46202 (dcrabb of page charges. The article must therefore be hereby marked “advertisement”
@iupui.edu). in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

http://www.ajpgi.org G1
Theme
G2 MOLECULAR MECHANISMS OF ALCOHOLIC FATTY LIVER

Fig. 1. Potential mechanisms underlying the


alcoholic fatty liver. Ethanol may influence
the activity of peroxisome proliferator-acti-
vated receptor-␣ (PPAR␣), sterol regulatory
element binding protein 1 (SREBP-1), and
AMP-dependent protein kinase (AMPK) di-
rectly or by way of effects on adiponectin
and TNF-␣. These effects in turn activate
lipogenic pathways, inhibit fatty acid oxida-
tion pathways, and increase the concentra-
tion of malonyl-CoA. Malonyl-CoA inhibits
the entry of free fatty acid (FFA) into the
mitochondria and blocks fatty acid oxidation
in the mitochondrion. CPT I, carnitine
palmitoyl-transferase; ACC, acetyl-CoA car-
boxylase; AdipoR2, adiponectin receptor 2;
TNF-␣ R1, TNF-␣ receptor 1.

fatty livers due to increased accumulation of cholesteryl esters and of hepatocytes, cells with the capability of oxidizing ethanol
triglycerides, and the fatty liver of obese (ob/ob) mice is caused by (7). This effect was not seen in cells that lack alcohol dehy-
elevated SREBP-1c levels, thereby increasing lipogenic gene drogenase (ADH), suggesting that metabolism of ethanol was
expression, enhancing fatty acid synthesis, and accelerating tri- required for this effect. Further support for this hypothesis was
glyceride accumulation in the liver. Recent work has shown that provided by showing that the effect of ethanol was abolished
ethanol feeding or exposure of cultured cells to ethanol leads to by the ADH inhibitor 4-methylpyrazole and augmented by the
increased levels of the mature form of SREBP-1 and induction of aldehyde dehydrogenase (ALDH) inhibitor cyanamide. Incu-
the mRNAs of many members of the lipogenic enzyme set. bation of cells with acetaldehyde (50 –200 ␮M) also reduced
Perhaps the most important questions remaining are: what the ability of PPAR␣ in nuclear extracts to bind DNA. Thus
mediates effects of ethanol on these transcription factors? Does acetaldehyde was likely responsible for the effect of ethanol.
ethanol affect a central regulatory system that is responsible for The activity of several other nuclear receptors (hepatocyte
both the reduction in PPAR␣ and the increase in SREBP-1 nuclear factor 4, apolipoprotein AI regulatory protein-1, and
activity, or are these the results of independent actions of chicken ovalbumin upstream promotor-transcription factor)
ethanol? Would prevention of fatty liver, even with continued was not affected by ethanol (7), suggesting that this is neither
alcohol consumption, prevent the more severe complications of nonspecific toxicity of acetaldehyde nor an effect on down-
alcoholic liver injury? Several new leads that may answer these stream signaling pathways shared by the nuclear receptors.
questions have appeared recently. Ethanol is reported to reduce
the blood level of adiponectin, a hormone made in the adipose EFFECTS OF ETHANOL ON PPAR␣ FUNCTION IN VIVO
tissue, which is reported to activate PPAR␣ and AMP-depen-
dent protein kinase (AMPK), and inhibit SREBP-1. Further- To determine whether this effect of ethanol also occurs in
more, AMPK has been reported to reduce the expression of vivo, chronic ethanol feeding studies were performed (6). Four
SREBP-1 and inhibit acetyl-CoA carboxylase (ACC). The weeks of ethanol feeding resulted in histological and biochem-
latter effect will reduce the level of malonyl-CoA and increase ical fat accumulation. PPAR␣ protein levels were relatively
the rate of entry of fatty acyl-CoA into the mitochondrion. This unchanged, whereas RXR␣ levels were substantially reduced.
review will explore the interactions of ethanol with these This may reflect the effect of increased portal vein endotoxin,
regulators of hepatic fat metabolism and the possibility of because reduced hepatic RXR␣ in response to endotoxin has
controlling fatty liver by pharmacological intervention. been reported (3). PPAR␣/RXR binding to its consensus se-
EFFECTS OF ETHANOL ON PPAR␣ FUNCTION IN
quence was also reduced significantly (6). Of the PPAR-
CULTURED CELLS
controlled gene battery, the mRNA for medium-chain acyl-
CoA dehydrogenase was decreased by 40%, but those encod-
Ethanol inhibited the transcriptional activating and DNA- ing acyl-CoA oxidase, CPT I, very-long-chain acyl-CoA
binding ability of the PPAR␣, and its response to ligands synthetase, and very-long-chain acyl-CoA dehydrogenase were
(clofibrate, WY14,643), in hepatoma cells or primary cultures unchanged by the ethanol feeding protocol. As reported by
AJP-Gastrointest Liver Physiol • VOL 287 • JULY 2004 • www.ajpgi.org
Theme
MOLECULAR MECHANISMS OF ALCOHOLIC FATTY LIVER G3
others, liver fatty acid binding protein was induced by ethanol edly. Furthermore, the amount of mature SREBP-1 increased
feeding. ⬃2.5-fold after H4IIEC3 cells were exposed to either ethanol
Treatment of the animals with the PPAR␣ agonist (50 mM) or acetaldehyde (200 ␮M). In agreement with our
WY14,643 alone for the last 2 wk of the experiment resulted in finding, a recent work showed that acetaldehyde treatment
induction of PPAR␣ protein ⬃1.5-fold, no change in RXR␣ enhanced the levels of the precursor and mature forms of
levels, a threefold increase in the binding of PPAR␣/RXR to its SREBP-1 in HepG2 cells in a dose-dependent manner, and the
consensus DNA sequence, and mRNA levels of many PPAR␣ enhancing effect increased over time, peaking at 24 and 48 h of
target genes (long-chain acyl-CoA dehydrogenase, medium- the exposure to acetaldehyde (13).
chain acyl-CoA dehydrogenase, acyl-coA oxidase, very-long-
chain acyl-CoA synthetase, and fatty acid binding protein) EFFECTS OF ETHANOL ON SREBP FUNCTION IN VIVO
significantly higher than in ethanol-treated mice.
We further investigated the effect of ethanol on SREBP-1 in
WY14,643 treatment of animals during the last 2 wk of
vivo by using the standard liquid diet, pair-feeding protocol.
ethanol feeding increased the level of PPAR␣ protein and its
Feeding mice a low-fat diet (4% safflower oil, 6% cocoa butter,
ability to bind DNA. It also induced mRNA levels of PPAR␣
and 72% high carbohydrate) with ethanol (27.5% of total
target genes, and this was associated with a higher rate of fatty
calories) for 4 wk led to fatty liver development. A substantial
acid ␤-oxidation, normalization of serum FFA and triglyceride increase in the amount of mature SREBP-1 protein was found
levels, and reversal of fat accumulation in the livers. Clearly, in the livers of ethanol-fed mice. Moreover, ethanol feeding
incomplete activation of the PPAR␣ battery of enzymes during increased mRNA expression of several known SREBP-1 target
ethanol consumption contributes to the development of alco- hepatic lipogenic genes including fatty acid synthase (FAS),
hol-induced fatty liver (6). steroyl-CoA desaturase, malic enzyme, ATP citrate lyase, and
Fatty acid oxidation is not solely controlled by the activity of ACC, indicating that ethanol-mediated induction of SREBP-1
enzymes of ␤-oxidation. The entry of fatty acyl-CoA into the maturation may be associated with the increase in expression
mitochondrion is controlled by the level of malonyl-CoA, of these genes (23). Consistent with our finding, Ji and
which is an allosteric regulator of CPT I. Malonyl-CoA is Kaplowitz (9) reported that feeding mice a high-fat diet (15.7%
synthesized by ACC and is degraded by MCD. The role of carbohydrate, 25% protein, 35% corn oil) with ethanol (24.3%)
PPAR␣ in regulating the expression and activity of the en- for 6 wk using the intragastric infusion model approximately
zymes involved in controlling malonyl-CoA levels has re- doubled SREBP-1 mRNA over 6 wk and increased the mature
ceived attention recently. Reduced rates of fatty acid oxidation form of SREBP-1 protein. Taken together, these studies sug-
in the hearts of PPAR␣ (⫺/⫺) mice were associated with gest that products of ethanol metabolism may increase hepatic
higher concentrations of malonyl-CoA and the reduced expres- lipogenesis through activating hepatic SREBP-1.
sion and activity of MCD, suggesting that PPAR␣ could
transcriptionally regulate expression of MCD (4). PPAR␣ INVOLVEMENT OF AMPK IN ETHANOL EFFECTS
activation (by WY14,643), but not PPAR␥ activation (by
troglitazone), increased MCD mRNA expression and activity AMPK is emerging as a “metabolic master switch” regulat-
in cardiac and skeletal muscle (24). Further studies demon- ing pathways of hepatic triglyceride and cholesterol synthesis.
strated that the promoter of MCD was activated 17-fold in cells It phosphorylates and inhibits enzymes involved in lipid me-
cotransfected with PPAR␣/RXR␣ expression plasmids, and tabolism such as 3-hydroxy-3-methyl glutamate-CoA reduc-
MCD mRNA levels in hepatoma cells and in the liver of tase and ACC. ACC is generally regarded as the rate-limiting
fenofibrate-fed rats were increased as well (12). Thus the enzyme in fatty acid biosynthesis, and as discussed, its product,
inhibition of PPAR␣ function by ethanol would likely also lead malonyl-CoA, is a potent inhibitor of CPT I. AMPK is the
to increased malonyl-CoA and inhibition of fatty acid entry major kinase responsible for the inactivation of ACC, and
into the mitochondrion. recently, the activation of MCD by AMPK was reported (15).
Therefore, activation of AMPK is expected to reduce malonyl-
EFFECTS OF ETHANOL ON SREBP FUNCTION IN CoA levels, increasing fatty acid oxidation.
CULTURED CELLS More interestingly, it has been firmly established that
SREBP-1 can be both transcriptionally and posttranscription-
The role of SREBPs in alcoholic fatty liver was first inves- ally regulated by AMPK. Several studies demonstrate that
tigated by our group examining the effect of ethanol on reduced SREBP-1 mRNA and protein expression in rat hepa-
transcription of an SRE-containing promoter in cultured hep- tocytes and liver result from AMPK activation. Activation of
atoma cells (23). Two rat hepatoma cell lines, H4IIEC3 and AMPK decreased the stability of mature SREBP-1 protein in
McA-RH7777, were transfected with the reporter pSyn SRE- hepatocytes by acceleration of its proteasomal degradation (25,
luciferase and treated with various concentrations of ethanol, 16). Therefore, we tested the hypothesis that AMPK may play
then they were harvested for assay of reporter enzymes. Eth- a role in mediating the effects of ethanol on SREBP-1. We
anol treatment significantly increased pSyn SRE reporter ac- found that metformin or aminoimidazole-4-carboxamide ribo-
tivities up to sixfold in these hepatoma cells. However, the nucleotide, two known AMPK activators, partially blocked the
same effect was not observed in CV-1 cells, again suggesting ethanol-mediated induction of SREBP-dependent promoter ac-
that acetaldehyde generated from ethanol metabolism might be tivity in hepatoma cells. It was further observed that chronic
required. This was again confirmed by the use of inhibitors of ethanol feeding inhibited hepatic AMPK activity and enhanced
ethanol metabolism. 4-Methylpyrazole nearly abolished the activity of its target protein ACC. Therefore, our findings
effect of ethanol on pSyn SRE-luciferase expression, whereas suggest that AMPK may play a role in regulating the effects of
cyanamide, the ALDH inhibitor, augmented the effect mark- ethanol on hepatic SREBP-1 activation, fatty acid metabolism,
AJP-Gastrointest Liver Physiol • VOL 287 • JULY 2004 • www.ajpgi.org
Theme
G4 MOLECULAR MECHANISMS OF ALCOHOLIC FATTY LIVER

and the development of alcoholic fatty liver. The potential role into these mice dramatically alleviated hepatomegaly, fatty
of AMPK in the development of alcoholic fatty liver was liver, and liver inflammation. Whereas chronic ethanol feeding
further suggested by a very recent report that the fat-derived significantly decreased the rate of hepatic fatty acid oxidation
hormone adiponectin, a known activator of AMPK, alleviated and reduced the activities of CPT I, ACC, and FAS, adiponec-
alcoholic fatty liver disease (20). tin treatment restored all these activities (20). Thus it is
possible that reduction of hepatic fatty acid oxidation during
ROLES OF TNF-␣ AND ADIPONECTIN IN ALCOHOLIC ethanol consumption results from the suppression of adiponec-
FATTY LIVER tin levels by ethanol. Ethanol could suppress adiponectin
through its activation of TNF-␣. Indeed, both hepatic expres-
The cytokine TNF-␣ has been suggested to play a pivotal sion and circulating levels of TNF-␣ increased with ethanol
role in the development of alcoholic liver injury. The evidence feeding, and adiponectin treatment markedly suppressed
for this comes largely from the intragastric alcohol feeding TNF-␣ production in these mice (20). This could partly explain
model. Depletion of Kupffer cells with gadolinium prevented the protective role of adiponectin against alcoholic fatty liver.
alcoholic liver injury; similarly, administration of neutralizing It is well known that adiponectin and TNF-␣ regulate each
antibody against TNF-␣ blocked liver injury (necrosis and other’s production and antagonize each other’s biological ef-
inflammation), with less effect on steatosis. The blockade of fects in their target tissues. The role of TNF-␣ in alcohol liver
TNF-␣ effects was not complete in this model, as evidenced by injury has been described above. Chronic ethanol consumption
elevation of transaminases in the ethanol-fed, antibody-treated led to increased circulating and local concentrations of TNF-␣
animals. In an effort to more definitively demonstrate the role in adipose tissue and liver. However, it remains unclear
of TNF-␣, TNF receptor knockout animals [lacking either TNF whether increased TNF-␣ resulting from ethanol feeding
receptor (TNFR)1 (p55) or TNFR1 (p75)] were fed ethanol by causes adiponectin reduction or whether suppression of adi-
the intragastric lavage technique. The animals lacking TNFR1 ponectin production by ethanol leads to TNF-␣ induction. This
(p55) had normal transaminases, negligible necrosis and in- issue is complex. A recent study (17) showed that moderate
flammation, and nearly absent steatosis (22). In addition, ad- alcohol consumption significantly increased plasma adiponec-
ministration of TNF-␣ to mice results in the development of tin levels both in healthy and relatively insulin-resistant mid-
fatty liver and has been linked to increased rates of fatty acid dle-aged men without affecting their plasma TNF-␣ levels.
synthesis and esterification and activation of SREBP (11). In Therefore, effects of ethanol on adiponectin expression appear
the systemic inflammatory state induced with endotoxin, to depend on the amount of alcohol consumed and the dietary
PPAR␣ expression is reported to be markedly reduced (18). context.
Thus it is interesting to speculate that the effects of ethanol on
both PPAR␣ and SREBP-1 might be mediated by increased DISCUSSION
portal endotoxin and Kupffer cell-generated TNF-␣. However,
an additional hormone, adiponectin, may participate in the The original biochemical explanation for alcoholic fatty
effects of ethanol. liver centered on the ability of ethanol metabolism to shift the
Adiponectin, also referred to as 30-kDa adipocyte comple- redox state of the liver and inhibit fatty acid oxidation. Sub-
ment-related protein (Acrp30), is a hormone secreted by adi- sequent studies found repression of some enzymes involved in
pocytes. Full-length adiponectin undergoes proteolytic pro- fatty acid oxidation and induction of lipogenic enzymes in
cessing, and a small amount of globular adiponectin (gAd) ethanol-fed animals, but there has been no unifying hypothesis
circulates in plasma. Two adiponectin receptors, AdipoR1 and to explain these effects. We suggest that the effects of ethanol
AdipoR2, were identified last year (21). AdipoR1 is a high- on lipid metabolism result from inhibition of PPAR␣ and
affinity receptor for globular adiponectin and a low-affinity stimulation of SREBP-1, in effect resulting in metabolic re-
receptor for the full-length ligand, whereas AdipoR2 is an modeling of the liver toward a fat-storing, rather than fat-
intermediate-affinity receptor for both forms of adiponectin. oxidizing, organ. These effects may, in turn, result from effects
AdipoR1 is abundantly expressed in skeletal muscle, whereas of ethanol on AMPK. It is attractive to view AMPK as a central
AdipoR2 is predominantly expressed in the liver; therefore, modulator of hepatic fat metabolism, especially because
only full-length adiponectin is active in the liver. A large body AMPK activity is subject to effects of ethanol on adiponectin
of work has shown that decreased levels of circulating adi- and possibly TNF-␣ (Fig. 1).
ponectin are associated with obesity, insulin resistance, Type 2 We suggest that proximal effects of ethanol include increas-
diabetes, and atherosclerosis, and administration of adiponectin ing portal vein endotoxin, adipose and Kupffer cell TNF-␣
diminished the abnormalities associated with the metabolic production, and a reduction in circulating adiponectin. Exactly
syndrome. Further studies revealed that the effect of adiponec- how ethanol affects the adipose tissue is uncertain. At the
tin is largely mediated by an increase in fatty acid oxidation hepatocyte, lower adiponectin would result in reduced activity
associated with activation of AMPK and PPAR␣ pathways of PPAR␣ and AMPK. The signal cascades between AdipoR2
downstream of the adiponectin receptors, both in vitro and in and these mediators are unknown. TNF-␣ is known to reduce
animals (16, 21). the expression of PPAR␣ in liver. The effects of TNF-␣ on
The potential role of adiponectin in alcoholic fatty liver was AMPK are not known and are a current area of interest in our
first suggested by a recent study (20). Chronic consumption of laboratory.
a high-fat, ethanol-containing diet significantly decreased cir- Inhibition of AMPK by ethanol feeding results in an increase
culating concentrations of adiponectin by 30 – 40% after 3– 4 in SREBP-1 activity. As a result, target genes for SREBP-1 are
wk and reduced adiponectin correlated closely with the devel- upregulated, contributing to increased hepatic lipid synthesis.
opment of liver injury. Delivery of recombinant adiponectin AMPK inhibition also results in increased activity of ACC and
AJP-Gastrointest Liver Physiol • VOL 287 • JULY 2004 • www.ajpgi.org
Theme
MOLECULAR MECHANISMS OF ALCOHOLIC FATTY LIVER G5
decreased activity of MCD by way of changes in the phos- ACKNOWLEDGMENTS
phorylation state of these enzymes and thus would reduce the We are indebted to the outstanding technical and intellectual contributions
rate of fatty acid oxidation via increased malonyl-CoA con- of Ruth Ann Ross, Yan Zeng, and Michinaga Matsumoto.
centrations. AMPK may also affect the activity of PPAR␣.
PPAR␣ is activated in heart by p38 MAP kinase (2), and the GRANTS
activity of p38 might be reduced by inhibition of AMPK This work was conducted with the support of National Institute on Alcohol
activity (19). Reduced PPAR␣ activity will lead to reduced Abuse and Alcoholism Grants AA-06434 (to D. W. Crabb), AA-013623 (to M.
capacity for fatty acid oxidation and reduced expression You), and P50-AA-07611 to the Indiana Alcohol Research Center.
of MCD.
REFERENCES
Whereas this sequence is attractive as an explanation for
ethanol effects in the intact animals, alternative actions of 1. Aoyama T, Peters JM, Iritani N, Nakajima T, Furihata K, Hashimoto
ethanol need to be kept in mind, particularly in light of the T, and Gonzales FJ. Altered constitutive expression of fatty acid-
metabolizing enzymes in mice lacking the peroxisome proliferator-acti-
ability of ethanol to inhibit PPAR␣ and activate SREBP-1 in vated receptor ␣ (PPAR␣). J Biol Chem 273: 5678 –5684, 1998.
cultured cells. Adiponectin cannot be playing a role in these 2. Barger PM, Browning AC, Garner AN, and Kelly DP. p38 mitogen-
experimental findings. Ethanol and acetaldehyde treatment are activated protein kinase activates peroxisome proliferator-activated recep-
reported to increase TNF-␣ production by many cells, includ- tor ␣: a potential role in the cardiac metabolic stress response. J Biol Chem
276: 44495– 44501, 2001.
ing hepatoma cells, so paracrine effects of TNF-␣ on AMPK, 3. Beigneux AP, Moser A H, Shigenaga,JK, Grunfeld C, and Feingold
PPAR␣, and SREBP-1 need to be further explored. Additional KR. The acute phase response is associated with retinoid X receptor
possibilities include formation of acetaldehyde-protein adducts repression in rodent liver. J Biol Chem 275: 16390 –16399, 2000.
with PPAR␣ or modulation of the function of PPAR␣ by 4. Campbell FM, Kozak R, Wagner A, Altarejos JY, Dyck JR, Belke DD,
Severson DL, Kelly DP, and Lopaschuk GD. A role for peroxisome
activation of yet other kinase cascades (for example, p42/44 proliferator-activated receptor ␣ (PPAR␣) in the control of cardiac mal-
MAPK, protein kinase A, p38 MAPK, protein kinase B/Akt, onyl-CoA levels: reduced fatty acid oxidation rates and increased glucose
and Stat5b have been reported to alter PPAR␣ activity in oxidation rates in the hearts of mice lacking PPAR␣ are associated with
various tissues). Ethanol might affect one of these pathways higher concentrations of malonyl-CoA and reduced expression of malo-
nyl-CoA decarboxylase. J Biol Chem 277: 4098 – 4103, 2002.
and thereby reduce the transcriptional activity PPAR␣. The 5. Costet P, Legendre C, More J, Edgar A, Galtier P, and Pineau T.
increased level of mature SREBP-1 might be stimulated by Peroxisome proliferator-activated receptor ␣-isoform deficiency leads to
effects of ethanol on the sterols that regulate activity of SREBP progressive dyslipidemia with sexually dimorphic obesity and steatosis.
cleavage activating protein (SCAP), the membrane milieu J Biol Chem 273: 29577–29585, 1998.
6. Fischer M, You M, Matsumoto M, and Crabb DW. Peroxisome
within which SCAP functions, the activity of the processing proliferator-activated receptor ␣ (PPAR␣) agonist treatment reverses
proteases S1P or S2P, or the rate of degradation of mature PPAR␣ dysfunction and abnormalities in hepatic lipid metabolism in
SREBP-1, which is believed to be a proteasomal process. ethanol-fed mice. J Biol Chem 278: 27997–28004, 2003.
The ability of compounds that activate PPAR␣ to reverse the 7. Galli A, Pinaire J, Fischer M, Dorris R, and Crabb DW. The tran-
scriptional activation of peroxisome proliferator-activated receptor ␣ is
development of fatty liver is conceptually important, because inhibited by ethanol metabolism: novel mechanism for the development of
in other models (e.g., feeding of saturated fat or medium-chain ethanol-induced fatty liver. J Biol Chem 276: 68 –75, 2001.
triglycerides, blocking the effect of TNF-␣ with antibodies), 8. Ip E, Farrell GC, Robertson G, Hall P, Kirsch R, and Leclercq I.
prevention of fatty liver was correlated with reduced overall Central role of PPAR␣-dependent hepatic lipid turnover in dietary steato-
injury to the liver. Exactly how these drugs effect this change hepatitis in mice. Hepatology 38: 123–132, 2003.
9. Ji C and Kaplowitz N. Betaine decreases hyperhomocysteinemia, endo-
is uncertain. The increased DNA binding activity of PPAR␣ plasmic reticulum stress, and liver injury in alcohol-fed mice. Gastroen-
that was seen in WY14,643-treated animals was associated terology 124: 1488 –1499, 2003.
with increased levels of mRNAs for a number of fat-metabo- 10. Kersten S, Seydoux J, Peters JM, Gonzalez FJ, Desvergne B, and
lizing enzymes, increased fatty acid oxidation capacity in liver Wahli W. Peroxisome proliferator-activated receptor ␣ mediates the
adaptive response to fasting. J Clin Invest 103: 1489 –1498, 1999.
homogenates, lowered plasma triglyceride and FFA levels, and 11. Lawler JF, Yin M, Diehl AM, Roberts E, and Chatterjee S. Tumor
increased plasma ketone body levels (6). These effects were necrosis factor ␣ stimulates the maturation of sterol regulatory element
observed even with ethanol in the diet. In our model, the binding protein 1 in human hepatocytes through the action of neutral
activity of ACC is central to the development of alcoholic fatty sphingomyelinase. J Biol Chem 273: 5053–5059, 1998.
12. Lee GY, Kim NH, Zhao ZS, Cha BS, and Kim YS. Peroxisomal
liver. The enzyme was induced by ethanol, possibly via proliferator-activated receptor ␣ activates transcription of the rat hepatic
SREBP-1 activation, and WY14,643 reduced its activity. Al- malonyl-CoA decarboxylase gene: A key regulation of malonyl-CoA
though ACC is not known to have a PPRE in its promoter, level. Biochem J 378: 983–990, 2004.
there is evidence that this enzyme is negatively regulated by 13. Lluis JM, Colell A, Garcia-Ruiz C, Kaplowitz N, and Fernandez-
Checa JC. Acetaldehyde impairs mitochondrial glutathione transport in
PPAR␣ agonists (14), possibly via activation of AMPK. It is HepG2 cells through endoplasmic reticulum stress. Gastroenterology 124:
thus uncertain whether the ability of fibrates to reverse fatty 708 –724, 2003.
liver depends mainly on inhibition of ACC and activation of 14. Munday MR and Hemingway CJ. The regulation of acetyl-CoA car-
MCD or whether induction of fatty acid-oxidizing enzymes is boxylase–a potential target for the action of hypolipidemic agents. Adv
Enzyme Regul 39: 205–234, 1999.
also required. Furthermore, the effect of fibrates on adiponectin 15. Ruderman NB, Park H, Kaushik VK, Dean D, Constant S, Prentki M,
and TNF-␣ needs to be evaluated. and Saha AK. AMPK as a metabolic switch in rat muscle, liver and
These studies on the genesis of fatty liver in alcohol-fed adipose tissue after exercise. Acta Physiol Scand 178: 435– 442, 2003.
animals lay the foundation for further clinical investigation. 16. Shklyaev S, Aslanidi G, Tennant M, Prima V, Kohlbrenner E, Krou-
tov V, Campbell-Thompson M, Crawford J, Shek EW, Scarpace PJ,
Modulation of these signaling pathways may provide novel and and Zolotukhin S. Sustained peripheral expression of transgene adi-
effective therapies for alcoholic fatty liver and possibly prevent ponectin offsets the development of diet-induced obesity in rats. Proc Natl
the inflammatory and fibrotic responses of the liver to ethanol. Acad Sci USA 100: 14217–14222, 2003.

AJP-Gastrointest Liver Physiol • VOL 287 • JULY 2004 • www.ajpgi.org


Theme
G6 MOLECULAR MECHANISMS OF ALCOHOLIC FATTY LIVER

17. Sierksma A, Patel H, Ouchi N, Kihara S, Funahashi T, Heine RJ, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T, Shimizu T, Nagai
Grobbee DE, Kluft C, and Hendriks HF. Effect of moderate alcohol R, and Kadowaki T. Cloning of adiponectin receptors that mediate
consumption on adiponectin, tumor necrosis factor-␣, and insulin sensi- antidiabetic metabolic effects. Nature 423: 762–769, 2003.
tivity. Diabetes Care 27: 184 –189, 2004. 22. Yin M, Wheeler MD, Kono H, Bradford BU, Gallucci RM, Luster MI,
18. Tai ES, bin Ali A, Zhang Q, Loh LM, Tan CE, Retnam L, El Oakley and Thurman RG. Essential role of tumor necrosis factor ␣ in alcohol-
RM, and Lim SK. Hepatic expression of PPAR␣, a molecular target of induced liver injury in mice. Gastroenterology 117: 942–952, 1999.
fibrates, is regulated during inflammation in a gender-specific manner. 23. You M, Fischer M, Deeg MA, and Crabb DW. Ethanol induces fatty
FEBS Lett 546: 237–240, 2003. acid synthesis pathways by activation of sterol regulatory element-binding
19. Xi X, Han J, and Zhang JZ. Stimulation of glucose transport by protein (SREBP). J Biol Chem 277: 29342–29347, 2002.
AMP-activated protein kinase via activation of p38 mitogen-activated 24. Young ME, Goodwin GW, Ying J, Guthrie P, Wilson CR, Laws FA,
protein kinase. J Biol Chem 276: 41029 – 41034, 2001. and Taegtmeyer H. Regulation of cardiac and skeletal muscle malonyl-
20. Xu A, Wang Y, Keshaw H, Xu LY, Lam KS, and Cooper GJ. The CoA decarboxylase by fatty acids. Am J Physiol Endocrinol Metab 280:
fat-derived hormone adiponectin alleviates alcoholic and nonalcoholic E471–E479, 2001.
fatty liver diseases in mice. J Clin Invest 112: 91–100, 2003. 25. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M,
21. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, Ventre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ,
Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K, Ohteki and Moller DE. Role of AMP-activated protein kinase in mechanism of
T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, Terauchi Y, metformin action. J Clin Invest 108: 1167–1174, 2001.

AJP-Gastrointest Liver Physiol • VOL 287 • JULY 2004 • www.ajpgi.org

You might also like