ArticlePDF AvailableLiterature Review

The role of fructose in the pathogenesis of NAFLD and the metabolic syndrome

Authors:

Abstract

Nonalcoholic fatty liver disease (NAFLD) is the most frequent liver disease worldwide, and is commonly associated with the metabolic syndrome. Secular trends in the prevalence of these diseases may be associated with the increased fructose consumption observed in the Western diet. NAFLD is characterized by two steps of liver injury: intrahepatic lipid accumulation (hepatic steatosis), and inflammatory progression to nonalcoholic steatohepatitis (NASH) (the 'two-hit' theory). In the first 'hit', hepatic metabolism of fructose promotes de novo lipogenesis and intrahepatic lipid, inhibition of mitochondrial beta-oxidation of long-chain fatty acids, triglyceride formation and steatosis, hepatic and skeletal muscle insulin resistance, and hyperglycemia. In the second 'hit', owing to the molecular instability of its five-membered furanose ring, fructose promotes protein fructosylation and formation of reactive oxygen species (ROS), which require quenching by hepatic antioxidants. Many patients with NASH also have micronutrient deficiencies and do not have enough antioxidant capacity to prevent synthesis of ROS, resulting in necroinflammation. We postulate that excessive dietary fructose consumption may underlie the development of NAFLD and the metabolic syndrome. Furthermore, we postulate that NAFLD and alcoholic fatty liver disease share the same pathogenesis.
NATURE REVIEWS
|
GASTROENTEROLOGY & HEPATOLOGY VOLUME 7
|
MAY 2010
|
251
Department of
Pediatrics, Korea
Cancer Center Hospital,
Gongneung-dong 215,
Nowon-gu, Seoul
139-706, Republic of
Korea (J. S. Lim).
Department of
Pediatrics, University of
California, San
Francisco,
513 Parnassus Avenue,
San Francisco,
CA 94143-0434, USA
(M. Mietus-Snyder,
A. Valente,
R. H. Lustig). College of
Osteopathic Medicine,
Touro University,
1310 Johnson Lane,
Mare Island, Vallejo,
CA 94592, USA
(J.-M. Schwarz).
Correspondence to:
R. H. Lustig
rlustig@peds.ucsf.edu
The role of fructose in the pathogenesis
of NAFLD and the metabolic syndrome
Jung Sub Lim, Michele Mietus-Snyder, Annie Valente, Jean-Marc Schwarz and Robert H. Lustig
Abstract | Nonalcoholic fatty liver disease (NAFLD) is the most frequent liver disease worldwide, and is
commonly associated with the metabolic syndrome. Secular trends in the prevalence of these diseases may
be associated with the increased fructose consumption observed in the Western diet. NAFLD is characterized
by two steps of liver injury: intrahepatic lipid accumulation (hepatic steatosis), and inflammatory progression
to nonalcoholic steatohepatitis (NASH) (the ‘two-hit’ theory). In the first ‘hit’, hepatic metabolism of fructose
promotes de novo lipogenesis and intrahepatic lipid, inhibition of mitochondrial β-oxidation of long-chain fatty
acids, triglyceride formation and steatosis, hepatic and skeletal muscle insulin resistance, and hyperglycemia.
In the second ‘hit’, owing to the molecular instability of its five-membered furanose ring, fructose promotes
protein fructosylation and formation of reactive oxygen species (ROS), which require quenching by hepatic
antioxidants. Many patients with NASH also have micronutrient deficiencies and do not have enough
antioxidant capacity to prevent synthesis of ROS, resulting in necroinflammation. We postulate that excessive
dietary fructose consumption may underlie the development of NAFLD and the metabolic syndrome.
Furthermore, we postulate that NAFLD and alcoholic fatty liver disease share the same pathogenesis.
Lim, J. S. et al. Nat. Rev. Gastroenterol. Hepatol. 7, 251–264 (2010); published online 6 April 2010; doi:10.1038/nrgastro.2010.41
Introduction
Nonalcoholic fatty liver disease (NAFLD) consists of a
benign form—hepatic steatosis (defined by the presence
of lipid droplets within hepatocytes on histopathologic
examination)—and the more perilous nonalcoholic
steato hepatitis (NASH), which may result in cirrhosis and
hepatic failure. NAFLD is currently the most common
liver disease worldwide, both in adults and children.1
Considering that NAFLD was first reported in adults
in 1980,2 and in children in 1983,3 the secular trend of
NAFLD prevalence is both astounding and alarming. In
the past 30 years, the prevalence and severity of NAFLD
has paralleled that of obesity, type 2 diabetes mellitus
(T2DM) and the metabolic syndrome, and a mech-
anistic association between NAFLD and these disorders
has been proposed.4 Despite concerted research into the
etiology and pathogenesis of NAFLD, the causes of
the disease remain unknown. Understanding the pathol-
ogy of NAFLD and other chronic metabolic diseases,
and how they relate to environmental changes that have
occurred in the past 30 years is essential to preventing
and treating NAFLD and the metabolic syndrome in
the future.
NAFLD and the metabolic syndrome
NAFLD is a condition of aberrant lipid storage in
hepatocytes.5 Although intrahepatic lipid accumulation
is related to lipid accumulation in adipocytes (that is,
obesity),6,7 not all intrahepatic lipid accumulation can
be explained by obesity alone. For instance, patients
with lipodystrophy are not obese, but have high levels of
intrahepatic lipid.8 Intrahepatic lipid accumulation is, in
fact, independent of BMI,9 but more accurately reflects
the existence of metabolic complications.10 The preva-
lence of the metabolic syndrome is 34% in American
adults overall, but 53% in patients with hepatic steato-
sis, and even higher (88%) in those with NASH.11 The
prevalence of both the metabolic syndrome and NAFLD
increases with age and with BMI; at autopsy, intra-
hepatic lipid is identified in 36% of lean adults and 72%
of adults with obesity,6 versus 5% of normal weight chil-
dren, 16% of overweight children, and 38% of children
with obesity.12
Patients with NAFLD are more likely to have dys-
lipidemia and increased blood pressure than individuals
of a similar weight without NAFLD.13,14 Furthermore,
the degree of intrahepatic lipid predicts metabolic dys-
function even better than does the degree of visceral
adipose tissue,15 and hepatic steatosis may precede the
onset of the metabolic syndrome and its complications.16
NAFLD more accurately predicts the existence of insulin
resistance than do the ATP III criteria, which were devel-
oped to diagnose the metabolic syndrome.17 In children18
and in adults,19 alanine aminotransferase (ALT) corre-
lates with both intrahepatic lipid, insulin resistance, and
other components of the metabolic syndrome.18 The
congruence of NAFLD and other manifesta tions of
the metabolic syndrome have led the Japanese govern-
ment to include a threshold level of ALT as part of their
definition of the metabolic syndrome.20
Competing interests
The authors declare no competing interests.
REVIEWS
nrgastro_41_MAY10.indd 251 15/4/10 12:08:53
© 20 Macmillan Publishers Limited. All rights reserved10
252
|
MAY 2010
|
voluMe 7 www.nature.com/nrgastro
Risk factors for NAFLD
Genetic risk factors
In a study of the correlation of ALT with the metabolic
syndrome, elevated levels of this enzyme were posi-
tively correlated with elevated serum lipid and glucose
concentra tions, blood pressure, and waist circumference
in most geographic regions except Asia,21 suggesting the
existence of hereditary modulators and/or alternate
pathogeneses of NAFLD in specific populations. NAFLD
affects specific racial and/or ethnic groups prefer entially22
and efforts have been made to find specific genetic pre-
dispositions for developing the disease. An associa-
tion between the presence of the rs738409 allele of the
patatin-like phospholipase domain-containing protein 3
(PNPLA3), which encodes a protein under metabolic
control, and intrahepatic lipid has been documented.
This allele is particularly frequent in Latino individuals,
who have the highest prevalence of NAFLD in the USA.22
Another allele in the same gene is associated with low
hepatic fat content in African Americans, who have the
lowest risk of developing NAFLD. The role of PNPLA3 in
lipid processing is not known, but this protein may also
affect other ectopic lipid depots, as visceral adipose tissue
is related to intrahepatic lipid accumulation, irrespective
of race or ethnicity.23
Metabolic risk factors
Obesity-associated insulin resistance and subsequent
hyperinsulinemia seem to be necessary though not suffi-
cient to precipitate the development of NAFLD.24 In a
retrospective study of children with biopsy-confirmed
NASH, 75% of them had fasting hyperinsulinemia,
which predicted steatosis, inflammation and fibrosis.25
Adolescents with both obesity and NAFLD had higher
liver and skeletal muscle insulin resistance, as measured
with hyperinsulinemic–euglycemic clamp techniques,
than those who were obese without NAFLD.13 Changes
in the concentrations of adipocytokines derived from
visceral adipose tissue, including increases in leptin,
resistin, tumor necrosis factor (TNF), and interleukin 6
concentrations, and decreases in adiponectin concentra-
tions, are all associated with NAFLD and insulin resis-
tance.26 Whether these changes in cytokine levels are
markers or causes of intrahepatic lipid, or whether all of
these factors are driven by an additional, yet unidentified
cause is not clear.
In addition, hyperglycemia may also exacerbate
NAFLD. In a Japanese study, the prevalence of steatosis
Key points
Nonalcoholic fatty liver disease (NAFLD) is commonly associated with the
metabolic syndrome
NAFLD can progress from a benign form (hepatic steatosis) to a more extreme
form (nonalcoholic steatoheaptitis)
Secular trends in fructose consumption coincide with those of NAFLD and the
metabolic syndrome; fructose is implicated in the pathogenesis of both NAFLD
and the metabolic syndrome
Hepatic fructose metabolism is reminiscent of that of ethanol; NAFLD and
alcoholic fatty liver disease are similar diseases
among lean adults was 62% in those who had been newly
diagnosed as having T2DM and 43% in those with
impaired fasting glucose levels, but only 27% in indivi duals
with normal fasting glucose levels.27
Dietary risk factors
Exposure to environmental factors, especially dietary
factors, is also likely to contribute to the generation of
intrahepatic lipid.28,29 Some studies have suggested that
specific dietary fats, such as trans-unsaturated fats,
contribute to hepatic steatosis.30,31 Conversely, mono-
unsaturated lipids such as oleic acid (the primary com-
ponent of olive oil),32 linoleic acid,33 or n–3 fatty acids34
decrease accumulation of intrahepatic lipid and improve
postprandial triglyceride levels, possibly by increasing
peroxisomal activity, which reduces damage by reac-
tive oxygen species (ROS).35 Another dietary factor that
probably contributes to hepatic steatosis, and the focus
of this Review, is the monosaccharide fructose. In case-
controlled studies, sugar-sweetened beverage consump-
tion was associated with hepatic steatosis, and this
association was independent of the degree of obesity.36,37
In other case-controlled studies, total fructose consump-
tion was associated with NAFLD in general, and NASH
in particular.38,39 Micro nutrient insufficiencies associated
with the consumption of sugar-sweetened beverages40
may aggravate their toxicity in producing NAFLD.
The ‘two-hit’ theory of NAFLD
Histologically, NAFLD is similar to alcoholic fatty liver
disease (AFLD). Mechanistically, although NAFLD is
thought to represent a continuum of hepatic insult, it
is probably the result of two distinct but related ‘hits’ to
the hepatocyte.41 These two steps of hepatic injury are
similar to those that are caused by ethanol and seen
in AFLD.42 The first ‘hit’ is the development of intra-
hepatic lipid and hepatic steatosis owing to an imbal-
ance of normal hepatic lipid metabolism, which results in
either excessive lipid influx, decreased lipid clearance, or
both. At this point, steatosis is potentially reversible and
does not necessarily lead to permanent hepatic injury.43
The second, less common, but more virulent ‘hit, which
occurs in 5% of individuals with steatosis, is a concomi-
tant inflammatory process that presumably results from
oxidative stress, lipid peroxidation and cytokine action.
The resulting lobular inflammation leads to ballooning
degeneration and perisinusoidal fibrosis, which promote
apoptosis and hepatocellular death, with resulting scar-
ring and progression to NASH.44 Approximately 25% of
patients with NASH will develop portal fibrosis, complet-
ing the process that leads to cirrhosis.45 Once inflamma-
tion has started, progression to cirrhosis may only take a
few years.46 Understanding the etiology of these two ‘hits’
is essential in the prevention and treatment of NAFLD.
The first ‘hit’: hepatic steatosis
Intrahepatic lipid accrues when the rate of hepatic lipid
influx, by fatty acid import or de novo synthesis of fatty
acids, exceeds the rate of hepatic lipid clearance, by
fatty acid catabolism or lipoprotein export (Figure 1).47,48
REVIEWS
nrgastro_41_MAY10.indd 252 15/4/10 12:08:53
© 20 Macmillan Publishers Limited. All rights reserved10
NATURE REVIEWS
|
GASTROENTEROLOGY & HEPATOLOGY VOLUM E 7
|
MAY 2010
|
253
The following five mechanisms that influence lipid influx
or clearance may lead to hepatic steatosis.
Increased ingestion of dietary fat
The role of dietary fat in the pathogenesis of NAFLD
remains controversial. Ad lib high-fat liquid feeding to
rats generates intrahepatic lipid,49 whereas ad lib feeding
of lipid-rich chow does not.50 In humans, ingestion of
dietary fat influences the accumulation of intrahepatic
lipid,51 but stable isotope studies have demonstrated
that up to only 15% of intrahepatic lipid is derived from
dietary fat.52 Lastly, low-carbohydrate diets, which are
otherwise rich in protein and fat, are frequently used as
treatment for NAFLD.53
Increased influx of free fatty acids
Free fatty acids that result from lipolysis of subcutaneous
or visceral adipose tissue depots circulate to the liver,
and may contribute to intrahepatic lipid accumula-
tion.54,55 For instance, in T2DM complicated by obesity
and chronic inflammation, overexpression of adipose
tissue cytokines can foment insulin resistance in the liver
and adipose tissue, in part by serine phosphorylation of
insulin receptor substrate 1 (IRS-1). Insulin resistance in
adipocytes leads to failure of insulin-mediated suppres-
sion of hormone- sensitive lipase (HSL) and release of
free fatty acids from adipose tissue (especially visceral
adipose) into the circulation.56 Release of free fatty acids
from visceral adipose tissue is particularly problem-
atic, as their first circulatory pass is through the liver.
Inflammation also increases TNF-mediated upregula-
tion of hepatic fatty acid translocase,57 which, coupled
with increased influx of free fatty acids, leads to steato-
sis.58 Other conditions characterized by increased lipo-
lysis and insulin resistance, however, do not necessarily
result in steatosis. For instance, patients with poorly con-
trolled type 1 diabetes mellitus have increased lipolysis
and insulin resistance, but have minimal intrahepatic
lipid accumulation,59 presumably because of enhanced
β-oxidation of fatty acids to ketones, which accelerates
hepatic lipid clearance for energy usage by the rest of
the body.
Increased de novo lipogenesis
De novo lipogenesis is accentuated more by excess
dietary carbohydrate than by excess dietary fat.60 For
example, when total energy intake from carbohydrate
exceeds total energy expenditure, hepatic de novo
lipogenesis is increased 10-fold.61 Similarly, the rate of
de novo lipo genesis associated with a high-carbohydrate
diet is 27-fold greater than that associated with a low-
carbohydrate diet in the fasting state, and fourfold greater
in the fed state.62
For example, patients with kwashiorkor (protein mal-
nutrition despite adequate caloric intake) who are fed
a maize diet, rich in carbohydrate,63 manifest severe
hepatic steatosis, more so than patients with other
malnutrition syndromes.64
Excess accumulation of metabolites generated by
de novo lipogenesis is seen in human and rat models
of hepatic steatosis.65,66 For instance, labeled isotope
studies in individuals with obesity and steatosis have
shown that 26.1% of the intrahepatic lipid pool is gen-
erated by de novo lipogenesis.52 On a typical high-fat
diet, lean individuals exhibited less than 3% (1–2 g per
day) of carbo hydrate converted to free fatty acids by
de novo lipogenesis,67,68 but this percentage was >10% in
indivi duals with obesity and insulin resistance.69
Impaired hepatic β-oxidation of fatty acids
β-Oxidation, (the sequential removal of two-carbon
fragments for ketone production or energy genera-
tion within mitochondria) is the main route for the
metabolism of long-chain fatty acids under normal
physiological conditions.70 This process is accomp-
lished by mitochondrial trifunctional protein, which
catalyzes three separate reactions to liberate these two-
carbon fragments from acyl-CoA, shortening them in
the process. This process, however, may be disrupted
at several key enzymatic stages. Dysfunction of this
protein may cause abrupt and massive hepatic failure
with steatosis,71 similar to the pathophysiology and
liver pathology in patients with Reye syndrome.72 The
transesterification and import of fatty acids into
the mitochondrial matrix for the process of β-oxidation
is achieved through covalent binding to carnitine, a
mitochondrial carrier molecule. Each fatty acid that is
transported requires the cleavage of the carnitine–fatty
acid complex, mediated by the enzyme carnitine
O-palmitoyltransferase 1 (CPT1). Regeneration of carni-
tine by CPT1 is the key rate-limiting and regulatory step
in the process of β-oxidation. Malonyl-CoA, formed by
dimerization and subsequent decarboxylation of acetyl-
CoA (the first step in fatty acid synthesis) is also a steric
Lipoprotein-
triglyceride
LDL receptor
Lipolysis
(T2DM, insulin
resistance)
De novo
lipogenesis
Reye
syndrome
Intrahepatic lipid
Lipid input Lipid output
VLDL-
triglyceride
Decreased VLDL
synthesis,
assembly,
secretion
Hypobetalipoproteinemia
β-oxidation
CO2
Ketones
Fructose
Carbohydrate
(Kwashiorkor)
Malonyl-CoA
Figure 1 | Pathways of hepatic lipid metabolism. Three main pathways contribute to
production of intrahepatic lipid (lipid input): import of lipoprotein triglyceride by the
LDL receptor; lipolysis, which is the source of free fatty acids that are imported
into the liver; and use of carbohydrate as an energy source by de novo lipogenesis.
Two pathways contribute to clearance of intrahepatic lipid (lipid output): complete
β-oxidation of intrahepatic lipid, which leads to carbon dioxide (CO2) production, or
incomplete β-oxidation, which leads to production of ketones (which are then
exported for energy use by the rest of the body); and lipid export, usually by
conjugating intrahepatic lipids with apolipoproteins to form VLDLs. Perturbation of
any of these pathways, such as in T2DM, kwashiorkor, Reye syndrome, or
hypobetalipoproteinemia, can result in accumulation of intrahepatic lipid and lead
to hepatic steatosis. Abbreviation: T2DM, type 2 diabetes mellitus.
REVIEWS
nrgastro_41_MAY10.indd 253 15/4/10 12:08:56
© 20 Macmillan Publishers Limited. All rights reserved10
254
|
MAY 2010
|
voluMe 7 www.nature.com/nrgastro
inhibitor of CPT1.73,74 Experimental suppression of
malonyl-CoA formation in rats counteracts the adverse
effects of NAFLD and insulin resistance.75 Malonyl-CoA
concentrations are increased when excessive citrate, the
primary substrate of de novo lipogenesis, is generated
beyond the oxidative capacity or needs of the liver; thus,
de novo lipogenesis and defective β-oxidation are linked,
and both promote intrahepatic lipid accumulation.
In lipid-engorged hepatocytes, several vicious cycles
involving TNF, ROS, peroxynitrite, and lipid peroxida-
tion products partially block the flow of electrons in
the respiratory chain. The over-reduction of upstream
respira tory chain complexes increases mitochondrial
ROS and peroxynitrite formation. Oxidative stress
increases the release of lipid peroxidation products
and cyto kines, which contribute to the development
of NASH.76 Impaired lipid β-oxidation seems to exert
only a minor influence in the development of NAFLD
in humans,77 but might arguably be enhanced in
persons with inadequate diet-derived and endogenous
anti oxidant defense mechanisms.78
Impaired triglyceride export
Hepatocytes esterify excess free fatty acids into tri-
glycerides, which are packaged with apolipoprotein
B-100 (apo B-100) by microsomal triglyceride transfer
protein (MTTP), and exported as VLDLs. VLDL pro-
duction in patients with NASH is decreased compared
with that in healthy controls,79 and this decrease could
accentuate the accumulation of triglycerides in the liver.
However, whether decreased VLDL production is a cause
or a result of NASH is not clear. Patients with hypo-
betalipoproteinemia—an autosomal recessive disease
caused by a defect in MTTP and characterized by fat
malabsorption, low plasma cholesterol levels at a young
age, and progressive neurologic degenerative disease—
have severe hepatic steatosis. Hepatic lipid export is
diminished in this condition, yet peripheral clearance
is normal, so affected individuals have markedly dimin-
ished serum triglyceride levels.80 Patients with NASH,
however, have hypertriglyceridemia due to impaired
clearance as well as defective lipid export.
Dietary factors and hepatic steatosis
Each of these five processes that control hepatic lipid
influx and/or clearance can be perturbed sufficiently
in humans to increase intrahepatic lipid and promote
hepatic steatosis, contributing to—but not fully explain-
ing—the global increase in prevalence and severity of
NAFLD. The inexorable rise in daily calorie intake seen
in the past 30 years in industrialized countries may also
have a role in this increase. In the USA, adult men have
increased caloric intake by 187 kcal per day, women
by 335 kcal per day, and adolescents of both sexes by
275 kcal per day.81 The prevalence of both NAFLD and
the metabolic syndrome have increased dramatically
worldwide with the global export of the Western diet.
Whether the excess of energy intake is the main factor
that underlies the NAFLD epidemic, whether specific
macronutrient and/or micronutrient components of the
diet are involved, or whether both factors have a role has
not been fully elucidated.
Dietary fat consumption
Despite documented increases in daily caloric intake,
total dietary fat intake has remained stable in the last
30 years (5.3 g per day decrease in men, 5 g per day
increase in women), while the percentage of calories
ingested from saturated fat has decreased.82 Although
high-fat feeding can induce overnutrition and intra-
hepatic lipid in experimental animal and human models,
it does so only with concomitant carbohydrate inges-
tion.50,51,83 Furthermore, long-term voluntary consump-
tion of an increased percentage of fat, as is seen in the
Atkins diet, did not result in increased caloric intake,
and did not increase the risk of obesity or NAFLD.84,85 In
a 2-year intervention study, participants on either the fat-
liberalized Mediterranean (monounsaturated fats) or the
Atkins (all fats) diet without imposed caloric restriction,
demonstrated comparable weight loss and decreased cir-
culating fasting insulin and triglyceride concentrations.86
Indeed, high-fat, low-carbohydrate diets seem to amelio-
rate elevated liver enzyme levels and decrease metabolic
disease risk.53 Therefore, the quantity of ingested dietary
fat does not seem to be the cause of NAFLD, although
qualitative fat composition may be of concern.
Trans-unsaturated fat consumption
With the advent of Crisco, the first commercial trans-
unsaturated or ‘partially hydrogenated’ fat, in 1911,
various processed food products, especially margarines
and baked goods, have included trans-unsaturated fat in
order to improve shelf life. Due to the trans- isomerization
of the double bond in these fatty acids, bacteria cannot
digest them, which prevents food from becoming rancid.
Unfortunately, our mitochondria (derived evolutionarily
from bacteria) also cannot digest trans-unsaturated
fats, which, therefore, do not undergo mitochondrial
β-oxidation,87 and increase the risk of hepatic steatosis.
Experimental feeding of trans-unsaturated fats (termed
the ‘Western Lifestyle Diet’) to animals is associated with
rapid accumulation of intrahepatic lipid.31 Consumption
of trans-unsaturated fats peaked in the 1960s, but since
1988, owing to the recognized association of trans-
unsaturated fat with cardiovascular disease, the percent-
age of calories from trans-unsaturated fat consumed in
the Western diet has been gradually decreasing. A reduc-
tion from 3.0% in 1982 to 2.2% in 2002 in total energy
derived from trans-unsaturated fats was documented in
the Minnesota Heart Survey.88 Thus, while consumption
of trans-unsaturated fats could mechanistically underlie
the development of NAFLD and the metabolic syndrome,
consumption trends are temporally disparate with the
current epidemic of these diseases.
Carbohydrate consumption
When it comes to which macronutrient promotes intra-
hepatic lipid, who knows better than the French? In
recog nition that dietary carbohydrate is more steato-
genic than fat, the prototypical fatty liver, ‘paté de foie
REVIEWS
nrgastro_41_MAY10.indd 254 15/4/10 12:08:56
© 20 Macmillan Publishers Limited. All rights reserved10
NATURE REVIEWS
|
GASTROENTEROLOGY & HEPATOLOGY VOLUM E 7
|
MAY 2010
|
255
gras, is made by force-feeding ducks or geese with a diet
of maize (corn), wheat, and soya cake—a macro nutrient
breakdown that favors carbohydrate (47.9%) over fat
(2.1%).89 In contrast to the results of dietary fat intake,
carbohydrate overfeeding in humans results in exces-
sive weight gain and hepatic steatosis in a short period of
time. For example, in the ‘Guru Walla’ overfeeding para-
digm, consumption of a diet of 7,000 kcal per day that
consists of 70% carbohydrate and 15% fat, and is mainly
composed by sorghum (a kind of maize), leads to hepatic
steatosis within a few weeks.90 Food consumption survey
data from the US Department of Agriculture, reported by
the National Health and Nutrition Examination Survey
program between 1971 and 2004, also indicate that the
observed increase in total energy intake in the US popula-
tion is accounted for almost completely by carbo hydrate
consumption, with a 67.7 g increase per day in men and
a 62.4 g increase per day in women within that time
frame.82 An excess in dietary carbohydrate consumption
has been reported specifically in patients with NASH.91
Secular trends in fructose intake
The predominant carbohydrate responsible for the rise in
caloric consumption associated with the typical Western
diet is the monosaccharide fructose, which is consumed
either as sucrose (50% fructose) or as high-fructose corn
syrup (42% or 55% fructose). Before 1900, US Americans
consumed approximately 15 g of fructose per day (4%
of total calories), mainly through fruits and vegetables.
Before World War II, fructose intake had increased to
24 g per day (5% of total calories); by 1977, it was 37 g
per day (7% of total calories); and by 1994, 55 g per day
(10% of total calories). Adolescents today consume over
72.8 g per day (12.1% of total calories) of fructose;92
20% of teenagers consume 25% or more of their total
calories as fructose.93 Thus, fructose consumption has
increased fivefold over the last century and more than
doubled in the last 30 years. Food disappearance data
from the Economic Research Service (ERS) of the US
Department of Agriculture support this secular trend.
Although the ERS documents partially decreased sucrose
intake per capita, the total annual consumption of caloric
sweeteners per capita has increased from 33 kg to 43 kg
in 30 years.94 Although the presence of high-fructose
corn syrup in soft drinks has received most of the atten-
tion,95,96 high fruit juice intake has also been associated
with childhood obesity.97 Currently, Americans consume
sugar at a rate of 66.8 kg per year (180 g per day), half of
which is fructose.
Fructose and the metabolic syndrome
Many investigators have implicated fructose in the
pathogenesis of the metabolic syndrome40,98–105 and
NAFLD.39,106,107 The liver is the principal site of fructose
metabolism, as it possesses the fructose-specific Glut5
transporter.108 Although adipo cytes possess GLUT-5
mRNA and protein, the level of this transporter in adipose
tissue is quite low.109 The kidney and small intestine also
possess GLUT-5 transporters, but their function is to
transport fructose molecules across their lumena, either
for urinary excretion (to eliminate any systemic fructose
that escapes hepatic clearance) or for release into the
portal circulation, which passes directly to the liver. The
hepatic metabolism of fructose is very different to that
of glucose in that it is insulin independent, bypasses the
process of glycolysis, and increases de novo lipo genesis
to a greater extent. Indeed, the hepatic metabolism of
fructose is more reminiscent of that of ethanol.110 Similar
to ethanol, fructose can induce each of the phenomena
associated with the metabolic syndrome (Figure 2).
Hypertension
Fructose is phosphorylated by fructokinase, which uses
ATP as the phosphate donor, depleting the hepatocyte of
intracellular ATP. The scavenger enzyme AMP deami-
nase 1 reclaims additional phosphates from ADP, and
in the process generates the waste product uric acid.
Uric acid acts within vascular smooth muscle to inhibit
endothelial nitric oxide synthase and resultant nitric
oxide production, which promotes hypertension.100
Our group has shown that sugar-sweetened beverage
consump tion positively correlates with uric acid and
blood pressure levels in children,111 while others have
documented this association in adults.112 Furthermore,
the uric acid inhibitor allopurinol can reduce blood
pressure in adolescents113 and adults with obesity.114
Hepatic steatosis
Owing to the excess substrate load, excess mitochondrial
acetyl-CoA is formed, exceeding the ability of the tri-
carboxylic acid (TCA) cycle to metabolize it. The excess
acetyl-CoA is converted to citrate, exits into the cytosol via
the citrate shuttle, and serves as the substrate for de novo
lipogenesis. Acetyl-CoA dimerizes and is decarboxy-
lated to form malonyl-CoA, which inhibits mitochon-
drial β-oxidation. Triglycerides newly formed by de novo
lipogenesis115 can overwhelm the lipid export machin-
ery and precipitate in the liver, forming intra hepatic
lipid and leading to hepatic steatosis.
Hepatic insulin resistance
Fructose-1-phosphate activates dual-specificity mitogen-
activated protein kinase kinase 7 (MKK7),116 which
stimulates the hepatic enzyme mitogen-activated protein
kinase 8 (MAPK8).117 This kinase is thought to be the
bridge between hepatic metabolism and inflamma-
tion.118 Furthermore, the intermediate diacylglycerol,
which accumulates during de novo lipogenesis acti-
vates hepatic protein kinase C ε type (PKCε).119 Both
MAPK8 and PKCε trigger serine phosphorylation and
subsequent inactivation of IRS-1, which leads to hepatic
insulin resistance.120–123
Dyslipidemia and muscle insulin resistance
Free fatty acids are also formed, which, when packaged as
triglycerides into heavily fat-laden VLDLs, are cleared with
low efficiency, causing dyslipidemia and augmenting the
risk of cardiovascular disease.115,124 Excess circulating lipid
is also taken up by skeletal muscle to form intra myocellular
lipid, which leads to muscle insulin resistance.125,126
REVIEWS
nrgastro_41_MAY10.indd 255 15/4/10 12:08:56
© 20 Macmillan Publishers Limited. All rights reserved10
256
|
MAY 2010
|
voluMe 7 www.nature.com/nrgastro
Hyperglycemia and T2DM
Fructose, a gluconeogenic precursor, increases synthe-
sis of the forkhead box protein O1 (FOXO1).127 Hepatic
insulin resistance, made worse by elevated fructose
concentra tions, prevents the phosphorylation of FOXO1,
which allows this protein to enter the nucleus and induce
the transcription of enzymes that promote gluconeo-
genesis. Increased hepatic glucose output foments hyper-
glycemia, and is likely to contribute to the development
of T2DM.
Obesity
Fructose also contributes to increased food consump-
tion and obesity. Direct effects of fructose on the central
nervous system (CNS) include stimulation of hormones
that stimulate appetite and reward,128–130 and reduction
of hypothalamic malonyl-CoA levels, which results in
increased AMP kinase concentrations, driving further
food intake.131 Indirect effects of fructose on the CNS
include hypertriglyceridemia, which reduces leptin
transport across the blood–brain barrier,132 and hyper-
insulinemia, which blocks the leptin signal transduction
pathway, resulting in a sense of starvation, again driving
further food intake.133
Endotoxinemia
Fructose consumption may also contribute to bacterial
overgrowth and increased intestinal permeability.39,105
Animal and human studies suggest that NAFLD is associ-
ated with small intestinal bacterial overgrowth,134 which
NO
Dyslipidemia
LPL
Leptin
resistance
Free fatty acids
Muscle
insulin
resistance
Triglycerides
Insulin
P
P
P
P
P
P
Uric acidIMP
PyruvatePyruvate
Liver
Mitochondria
Fructose-1,6-bis-P
Fructose-6-P
Glyceraldehyde
Dihydroxyacetone-P
PP2A
Xylulose-5-P
PGC-1β
PFK
Citrate
Fructose-1-P
Fructokinase ADP AMP
ATP
Pi
Blood pressure
Inammation
Hyperglycemia
Hepatic insulin
resistance
Glucose
Gluconeogenesis
Fructose
Fructose
GLUT-5
MKK7
MAPK8
DAG
Lipid
droplet
pSer-IRS-1
IRS-1
PKCε
ACLACC1 FAS
TCA
cycle
O2
ATP+
CO2
Acetyl-CoA
Acetyl-CoA Acyl-CoA VLDL
ApoB
MTTP
Malonyl-CoA
CPT-1
Citrate
ChREBP
AMP deaminase 1
Obesity
FOXO1
SREBP1c
Figure 2 | Hepatic fructose metabolism. Fructose induces: substrate-dependent phosphate depletion, which increases uric
acid and contributes to hypertension through inhibition of endothelial nitric oxide synthase and reduction of NO (green);
excess formation of citrate, which serves as the substrate for de novo lipogenesis (orange); excess formation of malonyl-
CoA, which inhibits β-oxidation (red); hepatic lipid droplet formation and steatosis; activation of MAPK8 and PKCε, which
contributes to serine phosphor ylation of IRS-1 and hepatic insulin resistance, which in turn promotes hyperinsulinemia and
influences substrate deposition into fat (yellow); export of free fatty acids, which leads to VLDL formation and muscle
insulin resistance (light blue); increased synthesis of FOXO1, which promotes gluconeogenesis and hyperglycemia (pink);
and central nervous system hyperinsulinemia, which antagonizes central leptin signaling and promotes continued energy
intake. Abbreviations: ACL, ATP-citrate lyase; ACC1, acetyl-CoA carboxylase 1; apo B, apolipoprotein B-100; ChREBP,
carbohydrate response element binding protein; CPT-1, carnitine O-palmitoyl transferase 1; FAS, fatty acid synthase; DAG,
diacylglycerol; FOXO1, forkhead box protein O1; GLUT-5, solute carrier family 2, facilitated glucose transporter member 5;
IRS-1, insulin receptor substrate 1; LPL, lipoprotein lipase; MAPK8, mitogen-activated protein kinase 8; MKK7, mitogen-
activated protein kinase kinase 7; MTTP, microsomal triglyceride transfer protein; NO, nitric oxide; PFK,
6-phosphofructokinase; PGC-1β, peroxisome proliferator-activated receptor γ coactivator 1β; Pi, inorganic phosphate; PKCε,
protein kinase C ε type; PP2A, protein phosphatase 2a; pSer-IRS-1, serine phosphorylated IRS-1; SREBP-1c, sterol
regulatory element binding protein-1c; TCA, tricarboxylic acid.
REVIEWS
nrgastro_41_MAY10.indd 256 15/4/10 12:08:58
© 20 Macmillan Publishers Limited. All rights reserved10
NATURE REVIEWS
|
GASTROENTEROLOGY & HEPATOLOGY VOLUM E 7
|
MAY 2010
|
257
is linked to circulating endotoxinemia and inflamma-
tory cytokines. The presence of these factors in the
blood has also been reported in patients with AFLD.135
Translocation of bacterial endotoxins through a leaky
gut lumen to the portal blood increases the exposure of
the liver to inflammation and injury. The gut microflora,
therefore, may be a key link between fructose feeding,
plasma endotoxin levels, and systemic and hepatic
inflammation associated with the metabolic syndrome.
Similarities between fructose and ethanol
Promotion of de novo lipogenesis
In contrast to the hepatic conversion of glucose pri-
marily to glycogen, a fructose bolus is metabolized by
glyco lysis directly to pyruvate (Figure 2). This process is
accentuated in individuals with insulin resistance and/or
obesity.52,69,136 Thus, in response to a fructose bolus, a large
volume of acetyl-CoA is generated to enter the hepatic
mitochondrial TCA cycle. This cycle, however, has a rela-
tively fixed maximum velocity to deal with its substrate,
modifiable by exercise, cold, altitude, and concentrations
of thyroid hormone. When the liver mitochondria are
not able to metabolize the entire fructose- derived acetyl-
CoA substrate excess, any extra exits the mitochondria
into the cytosol in the form of citrate via the citrate
shuttle.137 In the cytosol, carbohydrate response element
binding protein (ChREBP) activates the enzymes respon-
sible for de novo lipo genesis,138,139 which transforms
the citrate, through malonyl-CoA, to generate fatty
acyl-CoA. Furthermore, fructose-1-P stimulates per-
oxisome proliferator- activated receptor γ coactivator
(PGC-1β), a trans criptional co-activator for sterol regu-
latory element binding protein 1c (SREBP-1c). SREBP-1c
further increases the activity of the enzymes involved in
de novo lipogenesis, which leads to the generation of even
more fatty acyl-CoA.140 The majority of the fatty acyl-
CoA is packaged into VLDL for export, but a proportion
accumulates instead as lipid droplets in the hepatocyte.29
This double activation of de novo lipogenesis, mediated
through both the carbo hydrate (ChREBP) and lipogenic
(SREBP-1c) transcription factor pathways, seems to be
exclusive to fructose, and probably explains its ability to
generate intrahepatic lipid very rapidly, as it relies upon
a substrate-driven effect that does not need insulin
stimula tion. Qualitatively and quantitatively, the genera-
tion of acetyl-CoA, increased de novo lipo genesis,141 and
increased intrahepatic lipid formation142 that result from
excessive fructose intake are similar to those seen with
ethanol consumption.
Inhibition of β-oxidation
Fructose increases the intracytosolic citrate available for
de novo lipogenesis. The process of de novo lipogenesis
involves three enzymes: ATP-citrate lyase, which recon-
verts citrate to acetyl-CoA; acetyl-CoA carboxylase 1
(ACC1), which dimerizes and decarboxylates acetyl-
CoA into malonyl-CoA; and fatty acid synthase, which
adds further two-carbon fragments to malonyl-CoA to
elongate it to form fatty acyl-CoA. These two-carbon
fragments are generated in the mitochondria, after
regeneration of carnitine by CPT1 after transesterifica-
tion and import of fatty acids into the mitochondrial
matrix. Malonyl-CoA, which is generated by de novo
lipogenesis from carbohydrate, inhibits CPT174 and pre-
vents β-oxidation,143 which contributes to intrahepatic
lipid accumulation. In animal models, defective fatty
acid oxidation and hepatic insulin resistance result in
steatosis.144 These same phenomena occur in animals
when ethanol is the energy substrate.145,146 In ethanol-
fed rodents, β-oxidation is blocked by increased activity
of ACC1, which leads to increased levels of malonyl-CoA
and decreased activity of CPT1.147
Suppression of hepatic lipid export
The principal exit strategy of intrahepatic lipid is its
export from the liver as VLDL. The synthesis of VLDL
depends on MTTP for transfer of lipids to the apo B-100
protein, which is necessary for its correct folding before
export. Peroxisome proliferator-activated receptor α
(PPARα) regulates mitochondrial and peroxisomal fatty
acid oxida tion, and stimulates hepatic MTTP expression,
coordinating energy stores and facilitating the normal
excretion of hepatic lipid for peripheral processing.148
Both fructose149 and ethanol148,150 reduce hepatic PPARα
activity, which results in downregulation of MTTP.151
Hepatic tri glyceride availability is the major determi-
nant of VLDL secretion rate, but MTTP activity seems
to determine VLDL size, which determines the rate of
clearance of these molecules in the plasma.152 Elevated
circulating VLDL concentrations in a hamster model
of insulin resistance induced by high fructose feeding
was associated with VLDL over production;153,154 while
decreased clearance of tri glyceride-rich VLDL has been
demonstrated in fructose-fed rat models.151,155 Similarly,
suppression of MTTP activity by ethanol increases
VLDL production but reduces lipid export machin-
er y.156 Triglyceride-enriched VLDL levels are typically
100
Alanine aminotransferase (U/l)
Sugar-sweetened-beverage consumption (kcal per day)
African American (n
=
72)
r
=
0.20
P
=
0.099
White (n
=
118)
r
=
0.23
P
=
0.013
80
60
40
20
0
0 200 400 600 800 1,000 1,200 1,400
Figure 3 | Association between sugar-sweetened beverage consumption and serum
alanine aminotransferase in a population of children seeking obesity treatment at
the University of California, San Francisco.164 These two variables are more
strongly correlated in white children than in African American children.11
REVIEWS
nrgastro_41_MAY10.indd 257 15/4/10 12:08:59
© 20 Macmillan Publishers Limited. All rights reserved10
258
|
MAY 2010
|
voluMe 7 www.nature.com/nrgastro
elevated in patients with alcoholism.157 Treatment with
a PPARα agonist leads to upregulation of MTTP and
increased VLDL export and turnover, and alleviates
the intrahepatic lipid accumulation of both NAFLD158
and AFLD.148,150
Chronic ethanol159 and fructose160 feeding in rat and
hamster models respectively, are also associated with
increased intestinal delivery of apolipoprotein B-48
(apo B-48). Apo B-48 is a protein unique to chylo microns.
It is only generated in the small intestine, where an alter-
natively spliced stop codon truncates the apo B-100
mRNA transcript. Chylomicrons containing apo B-48
delivered to the liver are metabolized into triglyceride-
rich remnant lipoproteins that, unlike VLDLs (which
contain apo B-100) and their LDL metabolites, cannot
be cleared by the LDL cell-surface receptor that mediates
endo cytosis of cholesterol- rich apo-B-100-containing
lipoproteins in all nucleated cells. This process contrib-
utes to enhanced circulation time of triglyceride-rich
proteins, hyper triglyceridemia, and atherogenicity.
While the specific role of fructose or ethanol in human
apo B-48 synthesis has not been examined, increased
production of intestinally derived particles containing
apo B-48 occurs in humans with hyperinsulinemia and
insulin resistance.161
Intrahepatic lipid accumulation
In mouse models, fructose overfeeding results in rapid
development of intrahepatic lipid, hyper triglyceridemia,
and insulin resistance.162,163 Our group has shown similar
correlative and causative phenomena in humans.
Analysis of data from the Weight Assessment for Teen
and Child Health (WATCH) Clinic at the University of
California, San Francisco, demonstrated that daily sugar-
sweetened beverage consumption assessed by recall
(in kcal per day) correlated with ALT concentrations in
white children, although this correlation was smaller
in African American children164 perhaps owing to genetic
differences in modulators of lipid metabolism, such
as PNPLA3165 or PPARα (Figure 3).166 Although ALT
concentrations are an imperfect measure of NAFLD,167
serum levels of this protein correlate, nonetheless, with
intrahepatic lipid accumulation, especially in children.168
A similar relationship between consumption of sugar-
sweetened beverages and NAFLD has also been shown
in adults.33,36 Moreover, in a crossover, isocaloric feeding
study in adults performed by our group, in which diets
rich in fructose or complex carbohydrates were com-
pared, fructose feeding increased intrahepatic lipid
accumulation by 38% within 8 days, as measured by
magnetic resonance spectroscopy.169 These results indi-
cate that, even in an isocaloric state, fructose is more
likely to overwhelm the hepatic lipoprotein packaging
machinery than other dietary factors, which results in
accumulation of intrahepatic lipid, hepatic steatosis,
and elevation of ALT. These associative and mechanistic
data from animals and humans point to excessive fruc-
tose consumption as a proximate cause of NAFLD. Once
fructose-induced obesity and insulin resistance are estab-
lished, other factors, such as adipocytokines and intesti-
nal endotoxins, may also increase intrahepatic lipid levels
and exacerbate hepatic dysfunction.
The second ‘hit’: inflammation
Fructose is different from glucose in yet another way, in
that it can promote hepatocellular damage. Molecularly,
H
H
H
HOH
H
D-Glucose
(linear form)
α-D-Glucopyranose
(Haworth projection)
OH
CH2OH
CH2OH
HO
HO HO
C
1
1
2
22
3
33
444
5
55
6
6CH2OH
6
C
O
O
O
a
b
OH
C
C
C
C
CC
OH
OH
OH
H
HHH
H
HOH
OH
OH
H
H
CC
H
H
HOH
H
D-Fructose
(linear form)
α-D-Fructofuranose
(Haworth projection)
OH
CH2OH
CH2OH
HOH2C
HOH2C
CH2OH
HO
C
1
1
1
2
2
2
3
33
4
44
5
55
6
CH2OH
6
O
O
O
C
C
C
CC
OH
OH
HO H
Oxygen
H
HOH
OH
OH
H
HCC
H
6
1
Carbon
Hydrogen
Figure 4 | Molecular renditions of glucose and fructose. a | Glucose in the linear, chair (Haworth), and space-occupying
projections. b | Fructose in the linear, chair (Haworth), and space-occupying projections. In the linear form, both substrates
possess a reactive aldehyde or ketal moiety, which can bind nonenzymatically to freely available amino groups of proteins. At
body temperature and pH, however, the chair form of glucose predominates. This conformation is a glucopyranose
(6-membered ring), with equatorial hydroxyl groups and is molecularly stable, which limits its protein reactivity. The chair form
of fructose is a fructofuranose (5-membered ring) with two axial hydroxymethyl groups that exert allosteric and ionic forces
on the unstable furanose ring, which favors the linear form. Thus, at body temperature and pH, the majority of fructose exists
in the linear form and is more reactive to proteins than glucose.
REVIEWS
nrgastro_41_MAY10.indd 258 15/4/10 12:09:00
© 20 Macmillan Publishers Limited. All rights reserved10
NATURE REVIEWS
|
GASTROENTEROLOGY & HEPATOLOGY VOLUM E 7
|
MAY 2010
|
259
glucose is found in two steroisomeric forms: the linear
aldehyde form, and the gluco pyranose (6-membered
ring) form (Figure 4). The aldehyde form of glucose
is highly reactive with ε-amino groups of lysine. The
nonenzymatic exothermic reaction between these
factors leads to protein glycation,170 which is termed
the Maillard or ‘browning’ reaction; this is the reason
why bananas become brown with time. However, at
37 °C and pH 7.4, the ring form is molecularly stable
and nonreactive, owing to its 6-membered gluco-
pyranose and equatorial hydroxyl groups. In these
conditions, 80% of glucose is thought to remain in the
ring form. Fructose is also found in two stereoisomeric
forms: the linear ketal form, and the fructofuranose
(5-membered ring) form. The latter has two axial
(abutting) hydroxymethyl groups, which exert allo-
steric and ionic forces to the unstable furanose ring
and drive it toward the linear form. Thus, at body tem-
perature and pH, 80% of fructose is thought to exist
in the linear form, with a reactive ketal group. This
difference explains why nonenzymatic fructosyla-
tion is seven times more rapid than protein glyca-
tion with glucose as the substrate.171,172 Each protein
fructosylation reaction releases a superoxide radical
(Figure 5).173 Fructose generates 100 times more ROS
than glucose,174,175 which, if not quenched by an anti-
oxidant (in the case of liver, gluta thione), can promote
hepatocellular damage.
OH
OH
H
2
N
O
O
O
HS
OH
OH
H
2
N
O
O
O
HS
OH
OH
H
2
N
O
O
O
S
OH
OH
H
2
NO
O
O
S
HN NH
NH
HN
HN
HN
NH
NH
Reduced
glutathione
Oxidized
glutathione
H
HOH
H
Fructose
OH
CH2OH
CH2OH
HO
CO
C
C
C
OH
Schiff base
OH
CH2OH
CH2OH
O2
HO
HC N+
CH
HC
HC
OH
Heyns product
OH
CH2OH
CH2OH
HC NH
CO
HC
HC
Protein Protein
Acetaldehyde
adduct
O
CCH3
HN
Acetaldehyde
CCH3
O
H
Ethanol
HO CH2CH3
NADH NAD+
Lipid
protein
Peroxidation
Cell damage
Necroinammation
Protein
ADH1B
Figure 5 | Generation of reactive oxygen species by fructose or ethanol. Fructose first forms an intermediate Schiff base
with the ε-amino group of lysine, which then spontaneously hydrogenates to form an irreversible Heyns rearrangement
product (hydroxyamide linkage or fructose adduct), through the ‘Maillard reaction’. Each protein fructosylation generates
one superoxide radical (O2
), which must be quenched by an antioxidant, such as glutathione. Alternatively, ethanol is
metabolized by alcohol dehydrogenase 1B (ADH1B) to NADH and acetaldehyde. Acetaldehyde, through a similar Maillard
reaction, forms acetaldehyde adducts, with generation of superoxide radicals, which must also be quenched by
antioxidants. In the absence of adequate antioxidant capacity, production of reactive oxygen species leads to peroxidation,
hepatocellular damage, necroinflammation (nonalcoholic steatohepatosis), fibrosis, and ultimately cirrhosis.
REVIEWS
nrgastro_41_MAY10.indd 259 15/4/10 12:09:00
© 20 Macmillan Publishers Limited. All rights reserved10
260
|
MAY 2010
|
voluMe 7 www.nature.com/nrgastro
Although difficult to demonstrate in humans, this
pheno menon is easily recapitulated in vitro. In one
study in mouse lymphoma cells, fructose induced non-
enzymatic fructosylation and DNA damage, which
resulted in internucleosomal cleavage and apoptosis.176
In a study of hepato cytes in monolayer culture, incuba-
tion with fructose yielded no direct damage.177 However,
when these hepatocytes were preincubated with sub-
lethal doses of hydrogen peroxide, their ROS-quenching
ability was extinguished. Incubation with fructose was
as toxic as that with other organic aldehydes and caused
hepatocellular death. These experiments suggest that in
a susceptible redox environment fructose can act as a
direct hepatotoxin.
A similar phenomenon was also demonstrated
in vivo. The methionine-choline-deficient diet in rats
generates hepatic steatosis and NASH within 3 weeks.178
Methionine is the source of the methyl group for gluta-
thione synthesis, and choline is a building block of
phosphatidyl choline, a required component of VLDL
particle assembly.179 Deficiency in methionine and
choline promotes intrahepatic lipid accumulation
and ROS-mediated damage. Interestingly, if the energy
substrate for the methionine-choline-deficient diet is
starch (glucose), no intrahepatic lipid formation or
hepato celluar death ensues; however, if the substrate
is sucrose (glucose plus fructose), the liver undergoes
massive steato sis, apoptosis, necrosis, and fibrosis.180
These data suggest that large volumes of fructose, in
combination with micronutrient insufficiencies, can
impair glutathione and possibly other hepatic anti-
oxidant reserves,181 contributing to the second ‘hit’ of
hepatic damage in NAFLD, and the evolution of hepatic
steatosis to NASH. Although not yet demonstrated in
humans, this hypothesis is particularly attractive, as
several studies have shown that children with obesity,
especially sugar- sweetened beverage drinkers, despite
being over nourished are also micronutrient mal-
nourished.182,183 In this sense, hepatic inflammation
caused by fructose is again reminiscent of that caused by
alcoholism, in which micronutrient deficiencies are also
commonplace.184 The hepatic conversion of ethanol to
acetaldehyde by the enzyme alcohol dehydro genase 1B,
which results in the generation of glyco aldehyde radi-
cals, is akin to the conversion of fructose to carbo-
nyl metabolites.146 Glutathione-dependent hepatic
detoxifica tion processes are impaired under condi tions
of nutritional deficiency,185 and improved with provi-
sion of nutrients that serve as methyl donors.186 In the
absence of adequate nutritional substrate, we surmise
that these toxins generate a flux of ROS that can exceed
antioxidant reserves. Hepatic ROS may be generated
through other mechanisms as well, such as oxidation
of saturated fat.187 ROS formation, either directly from
fructose, ethanol or fat, or indirectly from mitochon-
drial dysfunction caused by defective β-oxidation, is a
likely initiator of endoplasmic reticulum stress106 and
the unfolded protein response,123,188 both of which may
also contribute to the pathogenesis of NAFLD and the
metabolic syndrome.189
Conclusions
Increased consumption of fructose, and the increase
in prevalence of obesity, the metabolic syndrome and
NAFLD in adults and children show no sign of plateau.
Every country that has adopted the fructose-rich, fiber-
poor, and micronutrient-poor Western diet has suffered
the same fate.190 Recognizing the relationship between
sugar consumption, cardiovascular disease, and the
metabolic syndrome, the American Heart Association
has recom mended reduction of daily sugar intake by
more than half.191 Other associations have called for
global fructose restriction.192 Elevated levels of fructose
have important effects on hepatic biochemistry and are
associated with cellular pathology. We postulate that
fructose is a proximate cause of both of the ‘hits’
that cause hepatic damage in NAFLD, recapitulating
the toxic effects of ethanol. This similarity is not sur-
prising, as fructose and ethanol are highly congruent
evolutionarily and biochemically, and ethanol is, in fact,
produced by fermentation of fructose. The only differ-
ence is that for fructose we humans have to perform
our own glycolysis, while for ethanol, yeast perform the
glycolysis for us. Excessive intake of both of these sub-
stances leads to excessive hepatic substrate burden, an
excess of mitochondrial acetyl-CoA being transformed
into citrate and shuttled to the cytosol, stimula tion of
de novo lipo genesis, and an excess of malonyl-CoA
inhibiting hepatic lipid β-oxidation. The combina-
tion of these factors is a recipe for hepatic steatosis.
The reactive aldehyde or ketal groups in both of these
molecules can lead to ROS formation, which, when
associated with lack of antioxidant capacity aggravated
by micronutrient mal nutrition, might cause necro-
inflammation, fibrosis, and subsequent cirrhosis. Given
these similarities, NAFLD and AFLD can be viewed as
similar diseases that are driven by ostensibly different
substrates. Similarly to alcoholism, we postulate that
disease prevention by reduction of fructose consump-
tion and improved nutritional status, rather than treat-
ment of NAFLD and the metabolic syndrome, is likely
to be the only rational public health effort to combat
these diseases.
Review criteria
Using PubMed and the substrate key terms “fructose”
or “ethanol”, combined with the effector terms “de novo
lipogenesis”, “hypertriglyceridemia”, “steatosis”, “fatty
liver”, “NAFLD”, “lipoproteins”, “insulin resistance”,
“metabolic syndrome”, and “reactive oxygen species”, a
review of the literature published between the years 1966
and 2009 on hepatic ethanol and fructose metabolism,
carbohydrate–protein adduct and reactive oxygen species
formation was conducted. Mechanistic studies in
animals that addressed directionality of effect, along with
correlative or mechanistic data in humans that supported
or detracted from such mechanisms were included. After
syntheses of these data, consultations with experts
in the field of fructose metabolism and hepatic lipid
metabolism were obtained to establish veracity of
these findings.
REVIEWS
nrgastro_41_MAY10.indd 260 15/4/10 12:09:00
© 20 Macmillan Publishers Limited. All rights reserved10
NATURE REVIEWS
|
GASTROENTEROLOGY & HEPATOLOGY VOLUM E 7
|
MAY 2010
|
261
1. Adams, L. A. & Lindor, K. D. Nonalcoholic fatty
liver disease. Ann. Epidemiol. 17, 863–869
(2007).
2. Ludwig, J., Viggiano, T. R., McGill, D. B. &
Ott, B. J. Nonalcoholic steatohepatitis:
Mayo clinic experience with a hither to unnamed
disease. Mayo Clin. Proc. 55, 434–438 (1980).
3. Moran, J. R., Ghishan, F. K., Halter, S. A. &
Greene, H. L. Steatohepatitis in obese children:
a cause of chronic liver dysfunction. Am. J.
Gastroenterol. 78, 374–377 (1983).
4. Roberts, E. A. Pediatric nonalcoholic fatty liver
disease (NAFLD): a “growing” problem?
J. Hepatol. 46, 1133–1142 (2007).
5. Unger, R. H., Clark, G. O., Scherer, P. E. & Orci, L.
Lipid homeostasis, lipotoxicity, and the
metabolic syndrome. Biochim. Biophys. Acta
1801, 209–214 (2010).
6. Wanless, I. R. & Lentz, J. S. Fatty liver hepatitis
(steatohepatitis) and obesity: an autopsy study
with analysis of risk factors. Hepatology 12,
1106–1110 (1990).
7. Kahn, C. R. Banting Lecture. Insulin action,
diabetogenes, and the cause of type II diabetes.
Diabetes 43, 1066–1084 (1994).
8. Javor, E. D. et al. Leptin reverses nonalcoholic
steatohepatitis in patients with severe
lipodystrophy. Hepatology 41, 753–760 (2005).
9. Seppala-Lindroos, A. et al. Fat accumulation in
the liver is associated with defects in insulin
suppression of glucose production and serum
free fatty acids independent of obesity in normal
men. J. Clin. Endocrinol. Metab. 87, 3023–3028
(2002).
10. Marchesini, G. et al. Association of nonalcoholic
fatty liver disease with insulin resistance. Am. J.
Med. 107, 450–455 (1999).
11. Marchesini, G. et al. Nonalcoholic fatty liver,
steatohepatitis, and the metabolic syndrome.
Hepatology 37, 917–923 (2003).
12. Schwimmer, J. B. et al. Prevalence of fatty liver in
children and adolescents. Pediatrics 118,
1388–1393 (2006).
13. Deivanayagam, S. et al. Nonalcoholic fatty liver
disease is associated with hepatic and skeletal
muscle insulin resistance in overweight
adolescents. Am. J. Clin. Nutr. 88, 257–262
(2008).
14. Le, K. A. & Bortolotti, M. Role of dietary
carbohydrates and macronutrients in the
pathogenesis of nonalcoholic fatty liver disease.
Curr. Opin. Clin. Nutr. Metab. Care 11, 477–482
(2009).
15. Fabbrini, E. et al. Intrahepatic fat, not visceral fat,
is linked with metabolic complications of obesity.
Proc. Natl Acad. Sci. USA 106, 15430–15435
(2009).
16. Fan, J. G. et al. Effects of nonalcoholic fatty liver
disease on the development of metabolic
disorders. J. Gastroenterol. Hepatol. 22,
1086–1091 (2007).
17. Musso, G. et al. Should nonalcoholic fatty liver
disease be included in the definition of
metabolic syndrome? A cross-sectional
comparison with Adult Treatment Panel III criteria
in nonobese nondiabetic subjects. Diabetes
Care 31, 562–568 (2008).
18. Burgert, T. S. et al. Alanine aminotransferase
levels and fatty liver in childhood obesity:
associations with insulin resistance,
adiponectin, and visceral fat. J. Clin. Endocrinol.
Metab. 91, 4287–4294 (2006).
19. Kotronen, A., Westerbacka, J., Bergholm, R.,
Pietiläinen, K. H. & Yki-Järvinen, H. Liver fat in
the metabolic syndrome. J. Clin. Endocrinol.
Metab. 92, 3490–3497 (2007).
20. Taki, K., Nishio, K., Hamajima, N. & Niwa, T.
Metabolic syndrome defined by new criteria in
Japanese is associated with increased liver
enzymes and C-reactive protein. Nagoya J. Med.
Sci. 70, 1–9 (2008).
21. Bethel, M. A. et al. Metabolic syndrome and
alanine aminotransferase: a global perspective
from the NAVIGATOR screening population.
Diabet. Med. 26, 1204–1211 (2009).
22. Romeo, S. et al. Genetic variation in PNPLA3
confers susceptibililty to nonalcoholic fatty liver
disease. Nat. Genet. 40, 1461–1465 (2008).
23. Kantartzis, K. et al. Dissociation between fatty
liver and insulin resistance in humans carrying a
variant of the patatin-like phospholipase 3 gene.
Diabetes 58, 2616–2623 (2009).
24. Pagano, G. et al. Nonalcoholic steatohepatitis,
insulin resistance, and metabolic syndrome:
further evidence for an etiologic association.
Hepatology 35, 367–372 (2002).
25. Schwimmer, J. B. et al. Obesity, insulin
resistance, and other clinicopathological
correlates of pediatric nonalcoholic fatty liver
disease. J. Pediatr. 143, 500–505 (2005).
26. Marra, F. & Bertolani, C. Adipokines in liver
diseases. Hepatology 50, 957–969 (2009).
27. Jimba, S. et al. Prevalence of non-alcoholic fatty
liver disease and its association with impaired
glucose metabolism in Japanese adults. Diabet.
Med. 22, 1141–1145 (2005).
28. Zivkovic, A. M., German, J. B. & Sanyal, A. J.
Comparative review of diets for the metabolic
syndrome: implications for nonalcoholic fatty
liver disease. Am. J. Clin. Nutr. 86, 285–300
(2007).
29. Cave, M. et al. Nonalcoholic fatty liver disease:
predisposing factors and the role of nutrition.
J. Nutr. Biochem. 18, 184–195 (2007).
30. Alkouri, N., Dixon, L. J. & Feldstein, A. E.
Lipotoxicity in nonalcoholic fatty liver disease:
not all lipids are created equal. Expert Rev.
Gastroenterol. Hepatol. 3, 445–451 (2009).
31. Tetri, L. H., Basaranoglu, M., Brunt, E. M.,
Yerian, L. M. & Neuschwander-Tetri, B. A. Severe
NAFLD with hepatic necroinflammatory changes
in mice fed trans fats and a high-fructose corn
syrup equivalent. Am. J. Physiol. Gastrointest.
Liver Physiol. 295, G987–G995 (2008).
32. Nagao, K., Inoue, N., Wang, Y. M., Shirouchi, B. &
Yanagita, T. Dietary conjugated linoleic acid
alleviates nonalcoholic fatty liver disease in
Zucker (fa/fa) rats. J. Nutr. 135, 9–13 (2005).
33. Assy, N., Nassar, F., Nasser, G. & Grosovski, M.
Olive oil consumption and non-alcoholic fatty
liver disease. World J. Gastroenterol. 15,
1809–1815 (2009).
34. Cussons, A. J., Watts, G. F., Mori, T. A. &
Stuckey, B. G. Omega-3 fatty acid
supplementation decreases liver fat content in
polycystic ovary syndrome: a randomized
controlled trial employing proton magnetic
resonance spectroscopy. J. Clin. Endocrinol.
Metab. 94, 3842–3848 (2009).
35. Zandbergen, F. & Plutzky, J. PPARalpha in
atherosclerosis and inflammation. Biochim.
Biophys. Acta 1771, 972–982 (2007).
36. Assy, N. et al. Soft drink consumption linked with
fatty liver in the absence of traditional risk
factors. Can. J. Gastroenterol. 22, 811–816
(2008).
37. Abid, A. et al. Soft drink consumption is
associated with fatty liver disease independent
of metabolic syndrome. J. Hepatol. 51, 918–924
(2009).
38. Ouyang, X. et al. Fructose consumption as a risk
factor for non-alcoholic fatty liver disease.
J. Hepatol. 48, 993–999 (2008).
39. Thuy, S. et al. Nonalcoholic fatty liver disease in
humans is associated with increased plasma
endotoxin and plasminogen activator inhibitor 1
concentrations and with fructose intake. J. Nutr.
138, 1452–1455 (2008).
40. Dhingra, R. et al. Soft drink consumption and
risk of developing cardiometabolic risk factors
and the metabolic syndrome in middle-aged
adults in the community. Circulation 116,
480–488 (2007).
41. James, O. F. & Day, C. P. Non-alcoholic
steatohepatitis (NASH): a disease of emerging
identity and importance. J. Hepatol. 29,
495–501 (1998).
42. Lieber, C. S. Alcoholic fatty liver : its
pathogenesis and mechanism of progression to
inflammation and fibrosis. Alcohol 34, 9–19
(2004).
43. Burt, A. D., Mutton, A. & Day, C. P. Diagnosis and
interpretation of steatosis and steatohepatitis.
Semin. Diagn. Pathol. 15, 246–258 (1998).
44. Syn, W. K., Choi, S. S. & Diehl, A. M. Apoptosis
and cytokines in non-alcoholic steatohepatitis.
Clin. Liver Dis. 13, 565–580 (2009).
45. Brunt, E. M. Histopathology of non-alcoholic fatty
liver disease. Clin. Liver Dis. 13, 533–544
(2009).
46. Patton, H. M. et al. Pediatric nonalcoholic fatty
liver disease: a critical appraisal of current data
and implications for future research. J. Pediatr.
Gastroenterol. Nutr. 43, 413–427 (2006).
47. Bradbur y, M. W. & Berk, P. D. Lipid metabolism in
hepatic steatosis. Clin. Liver Dis. 8, 639–671
(2004).
48. Koteish, A. & Diehl, A. M. Animal models of
steatosis. Semin. Liver Dis. 21, 89–104 (2001).
49. Lieber, C. S. et al. Model of nonalcoholic
steatohepatitis. Am. J. Clin. Nutr. 79, 502–509
(2004).
50. Romestaing, C. et al. Long term highly saturated
fat diet does not induce NASH in Wistar rats.
Nutr. Metab. (Lond.) 4, 4 (2007).
51. Westerbacka, J. et al. Dietary fat content
modifies liver fat in overweight nondiabetic
subjects. J. Clin. Endocrinol. Metab. 90,
2804–2809 (2005).
52. Donnelly, K. L. et al. Sources of fatty acids
stored in liver and secreted via lipoproteins in
patients with nonalcoholic fatty liver disease.
J. Clin. Invest. 115, 1343–1351 (2005).
53. York, L. W., Puthalapattu, S. & Wu, G. Y.
Nonalcoholic fatty liver disease and low-
carbohydrate diets. Ann. Rev. Nutr. 29, 365–379
(2009).
54. Anstee, Q. M. & Goldin, R. D. Mouse models in
non-alcoholic fatty liver disease and
steatohepatitis research. Int. J. Exp. Pathol. 87,
1–16 (2006).
55. Roden, M. Mechanisms of disease:
hepatic steatosis in type 2 diabetes-
pathogenesis and clinical relevance. Nat. Clin.
Pract. Endocrinol. Metab. 2, 335–348 (2006).
56. Shoelson, S. E., Lee, J. & Yuan, M. Inflammation
and the IKK beta/I kappa B/NF-kappa B axis in
obesity- and diet-induced insulin resistance. Int.
J. Obes. Relat. Metab. Disord. 27 (Suppl. 3),
S49–S52 (2003).
57. Memon, R. A., Feingold, K. R., Moser, A. H.,
Fuller, J. & Grunfeld, C. Regulation of fatty acid
transport protein and fatty acid translocase
mRNA levels by endotoxin and cytokines. Am. J.
Physiol. 274, E210–E217 (1998).
58. Ravikumar, B. et al. Real-time assessment of
postprandial fat storage in liver and skeletal
muscle in health and type 2 diabetes. Am. J.
Physiol. Endocrinol. Metab. 288, E789–E797
(2005).
59. Perseghin, G. et al. Reduced intrahepatic fat
content is associated with increased whole-body
lipid oxidation in patients with type 1 diabetes.
Diabetologia 48, 2615–2621 (2005).
REVIEWS
nrgastro_41_MAY10.indd 261 15/4/10 12:09:01
© 20 Macmillan Publishers Limited. All rights reserved10
262
|
MAY 2010
|
voluMe 7 www.nature.com/nrgastro
60. Schwarz, J. M., Neese, R. A., Turner, S., Dare, D.
& Hellerstein, M. K. Short-term alterations in
carbohydrate energy intake in humans. Striking
effects on hepatic glucose production, de novo
lipogenesis, lipolysis, and whole-body fuel
selection. J. Clin. Invest. 96, 2735–2743
(1995).
61. Aarsland, A., Chinkes, D. & Wolfe, R. R.
Contributions of de novo synthesis of fatty acids
to total VLDL-triglyceride secretion during
prolonged hyperglycemia/hyperinsulinemia in
normal man. J. Clin. Invest. 98, 2008–2017
(1996).
62. Hudgins, L. C. et al. Relationship between
carbohydrate-induced hypertriglyceridemia and
fatty acid synthesis in lean and obese subjects.
J. Lipid Res. 41, 595–604 (2000).
63. Williams, C. D., Oxon, B. M. & Lond, H.
Kwashiorkor: a nutritional disease of children
associated with a maize diet. 1935. Bull. World
Health Organ. 81, 912–913 (2003).
64. Doher ty, J. F., Adam, E. J., Griffin, G. E. &
Golden, M. H. Ultrasonographic assessment of
the extent of hepatic steatosis in severe
malnutrition. Arch. Dis. Child. 67, 1348–1352
(1992).
65. Shimomura, I., Bashmakov, Y. & Horton, J. D.
Increased levels of nuclear SREBP-1c
associated with fatty livers in two mouse
models of diabetes mellitus. J. Biol. Chem. 274,
30028–30032 (1999).
66. Araya, J. et al. Increase in long-chain
polyunsaturated fatty acid n - 6/n - 3 ratio in
relation to hepatic steatosis in patients with
non-alcoholic fatty liver disease. Clin. Sci. (Lond.)
106, 635–643 (2004).
67. Leitch, C. A. & Jones, P. J. Measurement of
human lipogenesis using deuterium
incorporation. J. Lipid Res. 34, 157–163 (1993).
68. Hellerstein, M. K. et al. Measurement of de novo
hepatic lipogenesis in humans using stable
isotopes. J. Clin. Invest. 87, 1841–1852 (1991).
69. Schwarz, J. M., Linfoot, P., Dare, D. &
Aghajanian, K. Hepatic de novo lipogenesis in
normoinsulinemic and hyperinsulinemic
subjects consuming high-fat, low-carbohydrate
and low-fat, high-carbohydrate isoenergetic
diets. Am. J. Clin. Nutr. 77, 43–50 (2003).
70. Glasgow, J. F., Middleton, B., Moore, R., Gray, A.
& Hill, J. The mechanism of inhibition of beta-
oxidation by aspirin metabolites in skin
fibroblasts from Reye’s syndrome patients and
controls. Biochim. Biophys. Acta 1454, 115–125
(1999).
71. Spiekerkoetter, U., Sun, B., Khuchua, Z.,
Bennett, M. J. & Strauss, A. W. Molecular and
phenotypic heterogeneity in mitochondrial
trifunctional protein de-ficiency due to beta-
subunit mutations. Hum. Mutat. 21, 598–607
(2003).
72. Barr, R., Glass, I. H. & Chawla, G. S. Reye’s
syndrome: massive fatty metamorphosis of the
liver with acute encephalopathy. Can. Med.
Assoc. J. 98, 1038–1044 (1968).
73. Schreurs, M., Kuipers, F. & van der Leij, F. R.
Regulatory enzymes of mitochondrial beta-
oxidation as targets for treatment of the
metabolic syndrome. Obes. Rev.
doi:10.1111/j.1467-789X.2009.00642.x.
74. McGarr y, J. D. & Brown, N. F. The mitochondrial
carnitine palmitoyltransferase system. From
concept to molecular analysis. Eur. J. Biochem.
244, 1–14 (1997).
75. Savage, D. B. et al. Reversal of diet-induced
hepatic steatosis and hepatic insulin resistance
by antisense oligonucleotide inhibitors of acetyl-
CoA carboxylases 1 and 2. J. Clin. Invest. 116,
817–824 (2006).
76. Pessayre, D. Role of mitochondria in non-
alcoholic fatty liver disease. J. Gastroenterol.
Hepatol. 22, S20–S27 (2007).
77. Kotronen, A. et al. Liver fat and lipid oxidation in
humans. Liver Int. 29, 1439–1446 (2009).
78. Auten, R. & Davis, J. Oxygen toxicity and reactive
oxygen species: the devil is in the details.
Pediatr. Res. 66, 121–127 (2009).
79. Charlton, M., Sreekumar, R., Rasmussen, D.,
Lindor, K. & Nair, K. S. Apolipoprotein synthesis
in nonalcoholic steatohepatitis. Hepatology 35,
898–904 (2002).
80. Berriot-Varoqueaux, N., Aggerbeck, L. P.,
Samson-Bouma, M. & Wetterau, J. R. The role of
the microsomal triglygeride transfer protein in
abetalipoproteinemia. Annu. Rev. Nutr. 20,
663–697 (2000).
81. [No auhtors listed] Trends in intake of energy
and macronutrients--United States, 1971–2000.
MMWR Morb. Mortal. Wkly Rep. 53, 80–82
(2004).
82. Chanmugam, P. et al. Did fat intake in the United
States really decline between 1989–1991 and
1994–1996? J. Am. Diet. Assoc. 103, 867–872
(2003).
83. Lar ter, C. Z. & Yeh, M. M. Animal models of
NASH: getting both pathology and metabolic
context right. J. Gastroenterol. Hepatol. 23,
1635–1648 (2008).
84. Stern, L. et al. The effects of low-carbohydrate
versus conventional weight loss diets in severely
obese adults: one-year follow-up of a randomized
trial. Ann. Intern. Med. 140, 778–785 (2004).
85. Samaha, F. F. et al. A low-carbohydrate as
compared with a low-fat diet in severe obesity.
N. Engl. J. Med. 348, 2074–2081 (2003).
86. Shai, I. et al. Weight loss with a low-
carbohydrate, Mediterranean, or low-fat diet.
N. Engl. J. Med. 359, 229–241 (2008).
87. Rasooly, R., Kelley, D. S., Greg, J. & Mackey, B. E.
Dietary trans 10, cis 12-conjugated linoleic acid
reduces the expression of fatty acid oxidation
and drug detoxification enzymes in mouse liver.
Br. J. Nutr. 97, 58–66 (2007).
88. Lee, S. et al. Trends in diet quality for coronary
heart disease prevention between 1980–1982
and 2000–2002: The Minnesota Heart Survey.
J. Am. Diet. Assoc. 107, 213–222 (2007).
89. European Commission. Welfare aspects of the
production of foie gras in ducks and geese
[online], http://ec.europa.eu/food/fs/sc/scah/
out17_en.pdf Ch. 4, 24–29 (2002).
90. Pasquet, P. et al. Massive overfeeding and energy
balance in men: the Guru Walla model. Am. J.
Clin. Nutr. 56, 483–490 (1992).
91. Toshimitsu, K. et al. Dietary habits and nutrient
intake in non-alcoholic steatohepatitis. Nutrition
23, 46–52 (2007).
92. Vos, M. B., Kimmons, J. E., Gillespie, C.,
Welsh, J. & Blanck, H. M. Dietary fructose
consumption among US children and adults: the
Third National Health and Nutrition Examination
Survey. Medscape J. Med. 10, 160 (2008).
93. Bray, G. A. How bad is fructose? Am. J. Clin. Nutr.
86, 895–896 (2007).
94. Haley, S., Reed, J., Lin, B.-H. & Cook, A. Sweetner
Consumption in the United States: Distribution
by Demographic and Product Characteristics.
Economic Research Service [online], http://
www.ers.usda.gov/Publications/SSS/Aug05/
SSS24301/ (2005).
95. Ludwig, D. S., Peterson, K. E. & Gortmaker, S. L.
Relation between consumption of sugar-
sweetened drinks and childhood obesity:
a prospective, obser vational analysis. Lancet
357, 505–508 (2001).
96. Warner, M. L., Harley, K., Bradman, A., Vargas, G.
& Eskenazi, B. Soda consumption and
overweight status of 2-year-old Mexican-
American children in California. Obesity 14,
1966–1974 (2006).
97. Faith, M. S., Dennison, B. A., Edmunds, L. S. &
Stratton, H. H. Fruit juice intake predicts
increased adiposity gain in children from low-
income families: weight status-by-environment
interaction. Pediatrics 118, 2066–2075 (2006).
98. Le, K. A. & Tappy, L. Metabolic effects of
fructose. Curr. Opin. Nutr. Metab. Care 9,
469–475 (2006).
99. Rutledge, A. C. & Adeli, K. Fructose and the
metabolic syndrome: pathophysiology and
molecular mechanisms. Nutr. Rev. 65, S13–S23
(2007).
100. Johnson, R. J. et al. Potential role of sugar
(fructose) in the epidemic of hypertension,
obesity and the metabolic syndrome, diabetes,
kidney disease, and cardiovascular disease. Am.
J. Clin. Nutr. 86, 899–906 (2007).
101. Havel, P. J. Dietary fructose: implications for
dysregulation of energy homeostasis and lipid/
carbohydrate metabolism. Nutr. Rev. 63,
133–157 (2005).
102. Gross, L. S., Li, S., Ford, E. S. & Liu, S. Increased
consumption of refined carbohydrates and the
epidemic of type 2 diabetes in the United
States: an ecologic assessment. Am. J. Clin.
Nutr. 79, 774–779 (2004).
103. Elliott, S. S., Keim, N. L., Stern, J. S., Teff, K. &
Havel, P. J. Fructose, weight gain, and the insulin
resistance syndrome. Am. J. Clin. Nutr. 76,
911–922 (2002).
104. Brown, C. M., Dulloo, A. G. & Montani, J. P.
Sugary drinks in the pathogenesis of obesity and
cardiovascular diseases. Int. J. Obes. 32,
528–534 (2008).
105. Spruss, A. & Bergheim, I. Dietary fructose and
intestinal barrier: potential risk factor in the
pathogenesis of nonalcoholic fatty liver disease.
J. Nutr. Biochem. 20, 657–662 (2009).
106. Collison, K. S. et al. Diabetes of the liver: the link
between nonalcoholic fatty liver disease and
HFCS-55. Obesity 17, 2003–2013 (2009).
107. Lê, K. A. et al. Fructose overconsumption causes
dyslipidemia and ectopic lipid deposition in
healthy subjects with and without a family
history of type 2 diabetes. Am. J. Clin. Nutr. 89,
1760–1765 (2009).
108. Drouard, V. & Ferraris, R. P. Regulation of the
fructose transporter Glut5 in health and
disease. Am. J. Physiol. Endocrinol. Metab. 295,
E227–E237 (2008).
109. Wood, I. S., Wang, B., Lorente-Cebrián, S. &
Trayhurn, P. Hypoxia increases expression of
selective facilitative glucose transporters (GLUT)
and 2-deoxy-D-glucose uptake in human
adipocytes. Biochem. Biophys. Res. Comm. 361,
468–473 (2007).
110. Lustig, R. H. The fructose epidemic. The
Bariatrician 24, 10–19 (2009).
111. Nguyen, S., Choi, H. K., Lustig, R. H. & Hsu, C. Y.
Sugar sweetened beverages, serum uric acid,
and blood pressure in adolescents J. Pediatr.
154, 807–813 (2009).
112. Gao, X. B. et al. Intake of added sugar and sugar-
sweetended drink and serum uric acid
concentration in U. S. men and women.
Hypertension 50, 306–312 (2007).
113. Feig, D. I., Soletsky, B. & Johnson, R. J. Effects of
allopurinol on blood pressure of adolescents
with newly diagnosed essential hypertension.
JAMA 300, 924–932 (2008).
114. Perez-Pozo, S. E. et al. Excessive fructose intake
induces the features of metabolic syndrome in
healthy adult men: role of uric acid in the
hypertensive response. Int. J. Obes.
doi:10.1038/ijo.2009.259.
REVIEWS
nrgastro_41_MAY10.indd 262 15/4/10 12:09:01
© 20 Macmillan Publishers Limited. All rights reserved10
NATURE REVIEWS
|
GASTROENTEROLOGY & HEPATOLOGY VOLUM E 7
|
MAY 2010
|
263
115. Stanhope, K. L. et al. Consuming fr uctose-, not
glucose-sweetened beverages increases
visceral adiposity and lipids and decreases
insulin sensitivity in overweight/obese humans.
J. Clin. Invest. 119, 1322–1334 (2009).
116. Wei, Y., Wang, D. & Pagliassotti, M. J. Fructose
selectively modulates c-jun N-terminal kinase
activity and insulin signaling in rat primary
hepatocytes. J. Nutr. 135, 1642–1646 (2005).
117. Wei, Y., Wang, D., Topczewski, F. &
Pagliassotti, M. J. Fructose-mediated stress
signaling in the liver: implications for hepatic
insulin resistance. J. Nutr. Biochem. 18, 1–9
(2007).
118. Hirosumi, J. et al. A central role for JNK in
obesity and insulin resistance. Nature 420,
333–336 (2002).
119. Morino, K., Petersen, K. F. & Shulman, G. I.
Molecular mechanisms of insulin resistance in
humans and their potential links with
mitochondrial dysfunction. Diabetes 55,
S9–S15 (2006).
120. Bezerra, R. M. N. et al. A high fructose diet
affects the early steps of insulin action in
muscle and liver of rats. J. Nutr. 130,
1531–1535 (2000).
121. Wei, Y. & Pagliassotti, M. J. Hepatospecific
effects of fructose on c-jun NH2-terminal
kinase: implications for hepatic insulin
resistance. Am. J. Physiol. Endocrinol. Metab.
287, E926–E933 (2004).
122. Tuncman, G. et al. Functional in vivo interactions
between JNK1 and JNK2 isoforms in obesity
and insulin resistance. Proc. Natl Acad. Sci. USA
103, 10741–10746 (2006).
123. Lim, J. H., Lee, H. J., Jung, M. H. & Song, J.
Coupling mitochondrial dysfunction to
endoplasmic reticulum stress response: a
molecular mechanism leading to hepatic insulin
resistance. Cell. Signal. 21, 169–177 (2008).
124. Teff, K. L. et al. Endocrine and metabolic effects
of consuming fructose- and glucose-sweetened
beverages with meals in obese men and
women: influence of insulin resistance on
plasma triglyceride responses. J. Clin.
Endocrinol. Metab. 94, 1562–1569 (2009).
125. Krssak, M. et al. Intramyocellular lipid
concentrations are correlated with insulin
sensitivity in humans: a 1H NMR spectroscopy
study. Diabetologia 42, 113–116 (1999).
126. Montell, E. et al. DAG accumulation from
saturated fatty acids desensitizes insulin
stimulation of glucose uptake in muscle cells.
Am. J. Physiol. Endocrinol. Metab. 280,
E229–E237 (2001).
127. Dong, X. C. et al. Inactivation of hepatic Foxo1
by insulin signaling is required for adaptive
nutrient homeostasis and endocrine growth
regulation. Cell Metab. 8, 65–76 (2008).
128. Lindqvist, A., Baelemans, A. & Erlanson-
Albertsson, C. Effects of sucrose, glucose and
fructose on peripheral and central appetite
signals. Regul. Pept. 150, 26–32 (2008).
129. Avena, N. M., Rada, P. & Hoebel, B. G. Evidence
for sugar addiction: behavioral and
neurochemical effects of intermittent, excessive
sugar intake. Neurosci. Biobehav. Rev. 32,
20–39 (2008).
130. Teff, K. L. et al. Dietary fructose reduces
circulating insulin and leptin, attenuates
postprandial suppression of ghrelin, and
increases triglycerides in women. J. Clin.
Endocrinol. Metab. 89, 2963–2972 (2004).
131. Cha, S. H., Wolfgang, M., Tokutake, Y.,
Chohnan, S. & Lane, M. D. Differential effects of
central fructose and glucose on hypothalamic
malonyl-CoA and food intake. Proc. Natl Acad.
Sci. USA 105, 16871–16875 (2008).
132. Banks, W. A. et al. Triglycerides induce leptin
resistance at the blood-brain barrier. Diabetes
53, 1253–1260 (2004).
133. Lustig, R. H. Childhood obesity: behavioral
aberration or biochemical drive? Reinterpreting
the first law of thermodynamics. Nat. Clin. Pract.
Endocrinol. Metab. 2, 447–458 (2006).
134. Sabaté, J. M. et al. High prevalence of small
intestinal bacterial overgrowth in patients with
morbid obesity: a contributor to severe hepatic
steatosis. Obes. Surg. 18, 371–377 (2008).
135. Rao, R. Endotoxemia and gut barrier dysfunction
in alcoholic liver disease. Hepatology 50,
638–644 (2009).
136. Faeh, D. et al. Effect of fructose overfeeding and
fish oil administration on hepatic de novo
lipogenesis and insulin sensitivity in healthy
men. Diabetes 54, 1907–1913 (2005).
137. Scott, C. C., Heckman, C. A. & Snyder, F.
Regulation of ether lipids and their precursors
in relation to glycolysis in cultured neoplastic
cells. Biochim. Biophys. Acta 575, 215–224
(1979).
138. Dentin, R. et al. Liver-specific inhibition of
ChREBP improves hepatic steatosis and insulin
resistance in ob/ob mice. Diabetes 55,
2159–2170 (2006).
139. Zivkovic, A. M., German, J. B. & Sanyal, A. J.
Comparative review of diets for the metabolic
syndrome: implications for nonalcoholic fatty
liver disease. Am. J. Clin. Nutr. 86, 285–300
(2007).
140. Nagai, Y. et al. The role of peroxisome
proliferator-activated receptor gamma
coactivator-1 beta in the pathogenesis of
fructose-induced insulin resistance. Cell Metab.
9, 252–264 (2009).
141. Siler, S. Q., Neese, R. A. & Hellerstein, M. K.
De novo lipogenesis, lipid kinetics, and whole-
body lipid balances in humans after acute
alcohol consumption. Am. J. Clin. Nutr. 70,
928–936 (1999).
142. You, M. & Crabb, D. W. Molecular mechanisms
of alcoholic fatty liver: role of sterol regulatory
element-binding proteins. Alcohol 34, 39–43
(2004).
143. Nagai, Y. et al. Amelioration of high fructose-
induced metabolic derangements by activation
of PPARalpha. Am. J. Physiol. Endocrinol. Metab.
282, E1180–E1190 (2002).
144. Zhang, D. et al. Mitochondrial dysfunction due to
long-chain Acyl-CoA dehydrogenase deficiency
causes hepatic steatosis and hepatic insulin
resistance. Proc. Natl Acad. Sci. USA 104,
17075–17080 (2007).
145. Guzmán, M. & Castro, J. Alterations in the
regulatory properties of hepatic fatty acid
oxidation and carnitine palmitoyltransferase I
activity after ethanol feeding and withdrawal.
Alcohol Clin. Exp. Res. 14, 472–477 (1990).
146. Farfán Labonne, B. E. et al. Acetaldehyde-
induced mitochondrial dysfunction sensitizes
hepatocytes to oxidative damage. Cell Biol.
Toxicol. 25, 599–609 (2009).
147. García-Villafranca, J., Guillén, A. & Castro, J.
Ethanol consumption impairs regulation of fatty
acid metabolism by decreasing the activity of
AMP activated protein kinase in rat liver.
Biochimie 90, 460–466 (2008).
148. Ameen, C. et al. Activation of peroxisome
proliferator-activated receptor alpha increases
the expression and activity of microsomal
triglyceride transfer protein in the liver. J. Biol.
Chem. 280, 1224–1229 (2005).
149. Roglans, N. et al. Impairment of hepatic Stat-3
activation and reduction of PPARalpha activity in
fructose-fed rats. Hepatology 45, 778–788
(2007).
150. Nanji, A. A., Dannenberg, A. J., Jokelainen, K. &
Bass, N. M. Alcoholic liver injury in the rat is
associated with reduced expression of
peroxisome proliferator-alpha (PPARalpha)-
regulated genes and is ameliorated by
PPARalpha activation. J. Pharmacol. Exp. Ther.
310, 417–424 (2004).
151. Koo, H. Y. et al. Dietary fructose induces a wide
range of genes with distinct shift in carbohydrate
and lipid metabolism in fed and fasted rat liver.
Biochim. Biophys. Acta 1782, 341–348 (2008).
152. Gambino, R., Cassader, M., Pagano, G.,
Durazzo, M. & Musso, G. Polymorphism in
microsomal triglyceride transfer protein: a link
between liver disease and atherogenic
postprandial lipid profile in NASH? Hepatology
45, 1097–1107 (2007).
153. Taghibiglou, C. et al. Hepatic very low density
lipoprotein-ApoB overproduction is associated
with attenuated hepatic insulin signaling and
overexpression of protein-tyrosine phosphatase
1B in a fructose fed hamster model of insulin
resistance. J. Biol. Chem. 277, 793–803 (2002).
154. Tsai, J. et al. Inflammator y NF-kappaB activation
promotes hepatic apolipoprotein B100
secretion: evidence for a link between hepatic
inflammation and lipoprotein production. Am. J.
Physiol. Gastrointest. Liver Physiol. 296,
1287–1298 (2009).
155. Hirano, T., Mamo, J. C., Poapst, M. E., Kuksis, A.
& Steiner, G. Impaired very low-density
lipoprotein-triglyceride catabolism in acute and
chronic fructose-fed rats. Am. J. Physiol. 256,
E559–E565 (1989).
156. Sozio, M. & Crabb, D. W. Alcohol and lipid
metabolism. Am. J. Physiol. Endocrinol. Metab.
295, E10–E16 (2008).
157. Steinberg, D., Pearson, T. A. & Kuller, L. H.
Alcohol and atherosclerosis. Ann. Intern. Med.
114, 967–976 (1991).
158. Svegliati-Baroni, G. et al. A model of insulin
resistance and nonalcoholic steatohepatitis in
rats: role of peroxisome proliferator-activated
receptor-alpha and n-3 polyunsaturated fatty
acid treatment on liver injury. Am. J. Pathol. 169,
846–860 (2006).
159. Lau, P. P., Cahill, D. J., Zhu, H. J. & Chan, L.
Ethanol modulates apolipoprotein B mRNA
editing in the rat. J. Lipid Res. 36, 2069–2078
(1995).
160. Haidari, M. et al. Fasting and postprandial
overproduction of intestinally derived
lipoproteins in an animal model of insulin
resistance. Evidence that chronic fructose
feeding in the hamster is accompanied by
enhanced intestinal de novo lipogenesis and
ApoB48-containing lipoprotein overproduction.
J. Biol. Chem. 277, 31646–31655 (2002).
161. Duez, H. et al. Hyperinsulinemia is associated
with increased production rate of intestinal
apolipoprotein B-48-containing lipoproteins in
humans. Arterioscler. Thromb. Vasc. Biol. 26,
1357–1363 (2006).
162. Jurgens, H. et al. Consuming fructose-sweetened
beverages increases body adiposity in mice.
Obes. Res. 13, 1146–1156 (2005).
163. Ackerman, Z. et al. Fructose-induced fatty liver
disease: hepatic effects of blood pressure and
plasma triglyceride reduction. Hypertension 45,
1012–1018 (2005).
164. Valente, A., Mietus-Snyder, M. L., Lim, J. S. &
Lustig, R. H. Association between sugar
sweetened beverage consumption and serum
alanini aminotransferase in obese children
[abstract]. Ped. Acad. Soc. 3854.45 (2009).
165. Romeo, S. et al. Genetic variation in PNPLA3
confers susceptibility to nonalcoholic fatty liver
disease. Nat. Genet. 40, 1461–1465 (2008).
REVIEWS
nrgastro_41_MAY10.indd 263 15/4/10 12:09:01
© 20 Macmillan Publishers Limited. All rights reserved10
264
|
MAY 2010
|
voluMe 7 www.nature.com/nrgastro
166. Shin, M. J., Kanaya, A. M. & Krauss, R. M.
Polymorphisms in the peroxisome proliferator
activated receptor alpha gene are associated
with levels of apolipoprotein CIII and triglyceride
in African-Americans but not Caucasians.
Atherosclerosis 198, 313–319 (2008).
167. Wieckowska, A. & Feldstein, A. E. Diagnosis of
nonalcoholic fatty liver disease: invasive versus
noninvasive. Semin. Liver Dis. 28, 386–395
(2008).
168. Radetti, G., Kleon, W., Stuefer, J. & Pittschieler, K.
Non-alcoholic fatty liver disease in obese children
evaluated by magnetic resonance imaging. Acta
Paediatr. 95, 833–837 (2006).
169. Schwarz, J. M. et al. Effects of short-term feeding
with high- vs low-fructose isoenergetic diets on
hepatic de novo lipogenesis, liver fat content and
glucose regulation [abstract]. Am. Diabetes
Assoc. 1476P (2009).
170. Dills, W. L. Protein fructosylation: fructose and
the Maillard reaction. Am. J. Clin. Nutr. 58,
779S–787S (1993).
171. Ahmed, N. & Fur th, A. J. Failure of common
glycation assays to detect glycation by fructose.
Clin. Chem. 38, 1301–1303 (1992).
172. Schalkwijk, C. G., Stehouwer, C. D. &
van Hinsbergh, V. W. Fructose-mediated non-
enzymatic glycation: sweet coupling or bad
modification. Diabetes Metab. Res. 20, 369–382
(2004).
173. Nagai, R. et al. Hydroxyl radical mediates
N epsilon-(carboxymethyl)lysine formation from
Amadori product. Biochem. Biophys. Res. Comm.
234, 167–172 (1997).
174. Bunn, H. F. & Higgins, P. J. Reaction of
monosaccharides with proteins: possible
evolutionary significance Science 213, 222–224
(1981).
175. Bose, T. & Chakraborti, A. S. Fructose-induced
structural and functional modifications of
hemoglobin: implication for oxidative stress in
diabetes mellitus. Biochim. Biophys. Acta 1780,
800–808 (2008).
176. Levi, B. & Werman, M. J. Fructose and related
phosphate derivatives impose DNA damage and
apoptosis in L5178Y mouse lymphoma cells.
J. Nutr. Biochem. 14, 49–60 (2003).
177. Lee, O. et al. Fructose and carbonyl metabolites
and endogenous toxins. Chem. Biol. Interact.
178, 332–339 (2009).
178. Koteish, A. & Diehl, A. M. Animal models of
steatohepatitis. Best Pract. Res. Clin.
Gastroenterol. 16, 679–690 (2002).
179. Zeisel, S. H. Dietary choline: biochemistry,
physiology, and pharmacology. Ann. Rev. Nutr. 1,
95–121 (1981).
180. Pickens, M. K. et al. Dietary sucrose is essential
to the development of liver injury in the MCD
model of steatohepatitis. J. Lipid Res. 50,
2072–2082 (2009).
181. Romestaing, C. et al. Mitochondrial adaptations
to steatohepatitis induced by a methionine- and
choline-deficient diet. Am. J. Physiol. Endocrinol.
Metab. 294, E110–E119 (2008).
182. Frary, C. D., Johnson, R. K. & Wang, M. Q.
Children and adolescents’ choices of foods and
beverages high in added sugars are associated
with intakes of key nutrients and food groups.
J. Adol. Health 34, 56–63 (2004).
183. Garcia, O. P., Long, K. Z. & Rosado, J. L. Impact
of micronutrient deficiencies on obesity. Nutr.
Rev. 67, 559–572 (2009).
184. Manari, A. P., Preedy, V. R. & Peters, T. J.
Nutritional intake of hazardous drinkers and
dependent alcoholics in the UK. Addict. Biol. 8,
201–210 (2003).
185. Godin, D. V. & Wohaieb, S. A. Nutritional
deficiency, starvation, and tissue antioxidant
status. Free Radic. Biol. Med. 5, 165–176
(1988).
186. Powell, C. L. et al. Mechanism for prevention of
alcohol-induced liver injury by dietar y methyl
donors. Toxicol. Sci. doi:10.1093/toxsci/
kfq031.
187. Gentile, C. L. & Pagliassotti, M. J. The role of
fatty acids in the development and progression
of nonalcoholic fatty liver disease. J. Nutr.
Biochem. 19, 567–576 (2008).
188. Santos, C. X., Tanaka, L. Y., Wosniak, J. &
Laurindo, F. R. Mechanisms and implications of
reactive oxygen species generation during the
unfolded protein response: roles of
endoplasmic reticulum oxidoreductases,
mitochondrial electron transport, and NADPH
oxidase. Antioxid. Redox Signal. 11, 2409–2427
(2009).
189. Kapoor, A. & Sanyal, A. J. Endoplasmic reticulum
stress and the unfolded protein response. Clin.
Liver Dis. 13, 581–590 (2009).
190. Lazo, M. & Clark, J. M. The epidemiology of
nonalcoholic fatty liver disease: a global
perspective. Semin. Liver Dis. 28, 339–350
(2008).
191. Johnson, R. K. et al. Dietary sugars intake and
cardiovascular health. A scientific statement
from the American Heart Association.
Circulation 120, 1011–1020 (2009).
192. Bantle, J. P. Dietary fructose and metabolic
syndrome and diabetes. J. Nutr. 139,
1263S–1268S (2009).
Acknowledgments
The authors would like to thank Drs S. Noworolski,
P. Tsai, P. Rosenthal, N. Bass, R. Merriman, and
R. Krauss for constructive input. Dr Schwarz’s
laboratory is supported by an NIH–National Institute
of Diabetes and Digestive and Kidney Disease grant
(R01 DK078133) and an American Diabetes
Association Clinical Research Award (1-08-CR-56).
REVIEWS
nrgastro_41_MAY10.indd 264 15/4/10 12:09:02
© 20 Macmillan Publishers Limited. All rights reserved10
... Our results showed that rats in the ORX + HFHF diet group had the highest degree of hepatic steatosis, lobular inflammation, hepatocyte ballooning, and percentage of positive Oil Red O staining area, including SREBP1c and FAS gene expression as compared to those of control, ORX + standard diet, and HFHF rats. Excessive consumption of fructose and saturated fat is associated with the buildup of liver fat, insulin resistance, and obesity, and they also contribute significantly to hepatic inflammation, thereby playing a key role in the development of NASH [29,30]. Fructose metabolism is recognized as a general pathway of hepatic de novo lipogenesis. ...
... The addition of fructose in the diet induces some transcriptional factors, i.e., carbohydrate responsive element binding protein (ChREBP) and SREBP1c, and also increases lipogenic gene expression when compared to HFD alone [31]. Additionally, fructose metabolism can account for ATP depletion, uric acid formation, oxidative stress production, and inflammation in the liver [29]. Since testosterone deficiency alone could increase de novo hepatic lipogenesis, it is not surprising that the combination of testosterone deficiency and HFHF diet consumption would lead to more severe hepatic steatosis and liver injury. ...
Article
Full-text available
Nonalcoholic steatohepatitis (NASH) is a progressive form of nonalcoholic fatty liver disease (NAFLD) that is characterized by hepatic inflammation and steatosis. Currently, limited data exist regarding the risk of NASH in transgender women and the treatment options for this particular population. The use of testosterone supplementation is unfavorable for transgender women, and estrogen supplementation is linked to an increased risk of breast cancer; thus, an isoflavone derivative compound known as “genistein” could serve as a viable substitute for a hormone supplement in this context. The purpose of this study was to investigate the treatment effects and mechanisms of actions of genistein and sex hormones in orchidectomized (ORX) rats with nonalcoholic steatohepatitis induced via a high-fat high-fructose diet (HFHF) model. Male Sprague-Dawley rats (n = 42) were randomly assigned into seven groups; control, ORX + standard diet, HFHF, ORX + HFHF, ORX + HFHF diet + testosterone (50 mg/kg body weight (BW) once weekly), ORX + HFHF diet + estradiol (1.6 mg/kg BW daily), and ORX + HFHF diet + genistein (16 mg/kg BW daily). The duration of the study was 6 weeks. Some parts of liver tissue were used for histological examination by H&E staining. The determination of fat accumulation was performed using Oil Red O staining. SREBP1c and FAS gene expression were quantified using real-time PCR technique. The levels of all types of peroxisome proliferator-activated receptors (PPARs; α, δ, γ), proteins, and signal transducer and activator of transcription 1 (STAT1) signaling pathway were determined by both immunoblotting and immunohistochemistry. Rats in the ORX + HFHF group had the highest degree of hepatic steatosis, lobular inflammation, and hepatocyte ballooning, and showed higher levels of genes related to de novo lipogenesis, including SREBP1c and FAS. The expression of PPARγ and STAT1 were upregulated, while the expression of PPARα and PPARδ were downregulated in the ORX + HFHF group. Testosterone, estradiol and genistein treatments improved NASH histopathology together with the reversal of all types of PPAR protein expressions. Interestingly, genistein decreased the levels of STAT1 protein expression more than those of testosterone and estradiol treatment. Genistein and sex hormone treatment could ameliorate NASH through the upregulation of PPARα, and PPARδ, and the suppression of PPARγ and STAT1 expression.
... A previous study showed that mice fed with fructose had a higher degree of hepatic lipid accumulation, plasma endotoxin levels and TNF-α protein expression than mice fed with other sugar types [22]. Moreover, fructose products could directly activate lipogenic genes, such as sterol-regulatory element binding protein (SREBP)-1c and carbohydrate-response element binding protein (ChREBP) and promote reactive oxygen species (ROS) overproduction and liver inflammation [23,24]. HDAC3 is a class I histone deacetylase that forms a complex with the nuclear hormone receptor co-repressors to regulate gene transcriptional repression. ...
Article
Full-text available
Testosterone deficiency has been reported to accelerate nonalcoholic fatty liver disease (NAFLD). However, there are minimal data on the risk of NAFLD in transgender women and the treatment of NAFLD in this population. This study aimed to investigate the treatment effects and the mechanisms of action of genistein and sex hormones in orchiectomized (ORX) rats with nonalcoholic steatohepatitis (NASH) induced by a high fat high fructose diet (HFHF). Seven-week old male Sprague-Dawley rats were randomly divided into 7 groups (n = 6 each group); 1) control group, 2) ORX + standard diet group, 3) HFHF group, 4) ORX + HFHF group, 5) ORX + HFHF diet + testosterone group (50 mg/kg body weight (BW) once weekly), 6) ORX + HFHF diet + estradiol group (1.6 mg/kg BW daily), and 7) ORX + HFHF diet + genistein group (16 mg/kg BW daily). The duration of treatment was 6 weeks. Liver tissue was used for histological examination by hematoxylin and eosin staining and hepatic fat measurement by Oil Red O staining. Protein expression levels of histone deacetylase3 (HDAC3) and peroxisome proliferator-activated receptor delta (PPARδ) were analyzed by immunoblotting. Hepatic nuclear factor (NF)-ĸB expression was evaluated by immunohistochemistry. Rats in the ORX + HFHF group had the highest degree of hepatic steatosis, lobular inflammation, hepatocyte ballooning and the highest percentage of positive Oil Red O staining area among all groups. The expression of HDAC3 and PPARδ was downregulated, while NF-ĸB expression was upregulated in the ORX + HFHF group when compared with control and ORX + standard diet groups. Testosterone, estradiol and genistein treatment improved histological features of NASH together with the reversal of HDAC3, PPARδ and NF-ĸB protein expression comparing with the ORX + HFHF group. In summary, genistein and sex hormone treatment could alleviate NASH through the up-regulation of HDAC3 and PPARδ, and the suppression of NF-ĸB expression.
Article
Scope: Fructose consumption from added sugars correlates with the epidemic rise in MetS and CVD. Maternal fructose intake has been described to program metabolic diseases in progeny. However, consumption of...
Article
Several models of mice-fed high-fat diets have been used to trigger non-alcoholic steatohepatitis and some chemical substances, such as carbon tetrachloride. This study aimed to evaluate the joint action of a high-fat diet and CCl4 in developing a short-term non-alcoholic steatohepatitis model. C57BL6/J mice were divided into two groups: standard diet-fed (SD), the high-fat diet-fed (HFD) and HFD + fructose-fed and carbon tetrachloride (HFD+CCl4). Animals fed with HFD+CCl4 presented increased lipid deposition compared with both SD and HFD mice. Plasma cholesterol was increased in animals from the HFD+CCl4 group compared to the SD and HFD groups, without significant differences between the SD and HFD groups. Plasma triglycerides showed no significant difference between the groups. The HFD+CCl4 animals had increased collagen deposition in the liver compared with both SD and HFD groups. Hydroxyproline was also increased in the HFD+CCl4 group. Liver enzymes, alanine aminotransferase and aspartate aminotransferase, were increased in the HFD+CCl4 group, compared with SD and HFD groups. Also, CCl4 was able to trigger an inflammatory process in the liver of HFD-fed animals by promoting an increase of ~2 times in macrophage activity, ~6 times in F4/80 gene expression, and pro-inflammatory cytokines (IL-1b and TNFa), in addition to an increase in inflammatory pathway protein phosphorylation (IKKbp). HFD e HFD+CCl4 animals increased glucose intolerance compared with SD mice, associated with reduced insulin-stimulated AKT activity in the liver. Therefore, our study has shown that short-term HFD feeding associated with fructose and CCl4 can trigger non-alcoholic steatohepatitis and cause damage to glucose metabolism.
Article
Full-text available
The zebrafish (Danio rerio) has emerged as an appreciated and versatile model organism for studying cardiovascular and metabolic diseases, offering unique advantages for both basic research and drug discovery. The genetic conservation between zebrafish and humans and their high fecundity and transparent embryos allow for efficient large-scale genetic and drug-oriented screening studies. Zebrafish possess a simplified cardiovascular system that shares similarities with mammals, making them particularly suitable for modeling various aspects of heart development, function, and disease. The transparency of zebrafish embryos enables the real-time visualization of cardiovascular dynamics, offering insights into early embryonic events and facilitating the study of heart-related anomalies. In metabolic research, zebrafish provide a cost-effective platform for modeling obesity, type 2 diabetes, hyperlipidemia, and other metabolic disorders. Their high reproductive rate allows for the generation of large cohorts for robust statistical analyses, while advanced genetic tools, such as CRISPR/Cas9, enable precise gene editing with which to model specific genetic mutations associated with human diseases. Zebrafish metabolic models have been instrumental in elucidating the molecular mechanisms underlying metabolic diseases, studying the effects of environmental factors, and identifying potential therapeutic targets. Additionally, the permeability of zebrafish embryos to small molecules facilitates drug discovery and screening, offering a rapid and economical approach to identifying compounds with therapeutic potential. In conclusion, zebrafish cardiovascular and metabolic disease models continue to contribute significantly to our perception of disease pathogenesis, providing a platform for translational research and developing novel therapeutic interventions. The versatility, scalability, and genetic manipulability of zebrafish position them as an invaluable asset in unraveling the complexities of cardiovascular and metabolic diseases. This review presents an overview of the zebrafish model’s key features and contributions to investigating cardiovascular and metabolic disorders. We discuss the benefits and drawbacks of using zebrafish models to study human disease and the critical findings revealed by the progress in this endeavor to date.
Article
Background Methylmalonic acidemia (MMA) is an autosomal recessive disorder caused by defects in propionyl‐CoA (P‐CoA) catabolism; of note, liver neoplasms rarely occur as a long‐term complication of the disorder. Herein, we report the case of a patient with MMA and hepatocellular carcinoma (HCC) who was successfully treated with a living‐donor liver transplant (LDLT) following prior kidney transplantation. Case Report A 25‐year‐old male patient with MMA underwent LDLT with a left lobe graft because of metabolic instability and liver neoplasms. He had presented with chronic symptoms of MMA, which had been diagnosed by genetic testing. Additionally, he had undergone living‐donor kidney transplantation with his father as the donor due to end‐stage kidney disease 6 years before the LDLT. He had an episode of metabolic decompensation triggered by coronavirus disease in 2019. Imaging studies revealed an intrahepatic neoplasm in the right hepatic lobe. Due to concerns about metabolic decompensation after hepatectomy, LDLT was performed using a left lobe graft obtained from the patient's mother. Pathological findings were consistent with the characteristics of well‐to‐moderately differentiated HCC. The postoperative course was uneventful, and the patient was discharged 48 days after the LDLT without any complications. At the 9‐month follow‐up, the patient's condition was satisfactory, with sufficient liver graft function and without metabolic decompensation. Conclusion This case indicates that although HCC is a rare complication in patients with MMA, clinicians should be aware of hepatic malignancies during long‐term follow‐up.
Article
Full-text available
Little is known about dietary habits and their relationships with liver disease in nonalcoholic fatty liver disease (NAFLD) patients, particularly in the absence of obesity, diabetes or hyperlipidemia. OBJECTIVE: To assess the association between soft drink consumption and the presence of fatty liver in NAFLD patients who do not have classic risk factors. METHODS: Three hundred ten patients with NAFLD diagnosed by ultrasound were assessed for 36 months in a cross-sectional manner. Thirty-one patients (10%) who had NAFLD without classic risk factors were compared with 30 healthy controls. Physical activity was assessed during the preceding week and year, and every six months for 36 months. Data on daily dietary intake of food and soft drink, and the source of added sugar were collected during two seven-day periods, at the beginning of the study, and within two weeks after the metabolic tests by using a validated food questionnaire given by a trained dietician. Insulin resistance and lipid peroxidation were assessed by homeostasis model assessment-insulin resistance index (HOMA-IRI) and malondialdehyde (MDA) levels, respectively. RESULTS: Eighty per cent of patients (25 of 31) consumed an excessive amount of soft drink beverages (more than 50 g/day of added sugar) for 36 months, compared with 20% in healthy controls (P
Article
During 1971-2000, the prevalence of obesity in the United States increased from 14.5% to 30.9%.¹ Unhealthy diets and sedentary behaviors have been identified as the primary causes of deaths attributable to obesity.² Evaluating trends in dietary intake is an important step in understanding the factors that contribute to the increase in obesity. To assess trends in intake of energy (i.e., kilocalories [kcals]), protein, carbohydrate, total fat, and saturated fat during 1971-2000, CDC analyzed data from four National Health and Nutrition Examination Surveys (NHANES): NHANES I (conducted during 1971-1974), NHANES II (1976-1980), NHANES III (1988-1994), and NHANES 1999-2000. This report summarizes the results of that analysis, which indicate that, during 1971-2000, mean energy intake in kcals increased, mean percentage of kcals from carbohydrate increased, and mean percentage of kcals from total fat and saturated fat decreased (Figures 1 and 2). An expert advisory committee appointed by the U.S. Department of Health and Human Services and the U.S. Department of Agriculture (USDA) is conducting a review of the Dietary Guidelines for Americans.³ Revised guidelines will be published in 2005.
Article
Background: A previous paper reported the 6-month comparison of weight loss and metabolic changes in obese adults randomly assigned to either a low-carbohydrate diet or a conventional weight loss diet. Objective: To review the 1-year outcomes between these diets. Design: Randomized trial. Setting: Philadelphia Veterans Affairs Medical Center. Participants: 132 obese adults with a body mass index of 35 kg/m 2 or greater; 83% had diabetes or the metabolic syndrome. Intervention: Participants received counseling to either restrict carbohydrate intake to <30 g per day (low-carbohydrate diet) or to restrict caloric intake by 500 calories per day with <30% of calories from fat (conventional diet). Measurements: Changes in weight, lipid levels, glycemic control, and insulin sensitivity. Results: By 1 year, mean (±SD) weight change for persons on the low-carbohydrate diet was -5.1 ± 8.7 kg compared with -3.1 ± 8.4 kg for persons on the conventional diet. Differences between groups were not significant (-1.9 kg [95% Cl, -4.9 to 1.0 kg]; P = 0.20). For persons on the low-carbohydrate diet, triglyceride levels decreased more (P = 0.044) and high-density lipoprotein cholesterol levels decreased less (P = 0.025). As seen in the small group of persons with diabetes (n = 54) and after adjustment for covariates, hemoglobin A 1c levels improved more for persons on the low-carbohydrate diet These more favorable metabolic responses to a low-carbohydrate diet remained significant after adjustment for weight loss differences. Changes in other lipids or insulin sensitivity did not differ between groups. Limitations: These findings are limited by a high dropout rate (34%) and by suboptimal dietary adherence of the enrolled persons. Conclusion: Participants on a low-carbohydrate diet had more favorable overall outcomes at 1 year than did those on a conventional diet. Weight loss was similar between groups, but effects on atherogenic dyslipidemia and glycemic control were still more favorable with a low-carbohydrate diet after adjustment for differences in weight loss.
Article
F ructose consumption (as both high fructose corn syr-up and sucrose) has increased coincidentally with the worldwide epidemics of obesity and metabolic syndrome. Fructose is a primary contributor to human disease as it is metabolized in the liver differently to glucose, and is more akin to that of ethanol. When consumed in large amounts, fructose promotes the same dose-dependent toxic effects as ethanol, promoting hypertension, hepatic and skeletal muscle insulin resistance, dyslipidemia and fatty liver disease. Also similar to ethanol, through di-rect stimulation of the central nervous system "hedonic pathway" and indirect stimulation of the "starvation path-way," fructose induces alterations in central nervous sys-tem energy signaling that lead to a vicious cycle of exces-sive consumption, with resultant morbidity and mortality. Fructose from any source should be regarded as "alcohol without the buzz." Obesity preven-tion and treatment is ineffective in the face of the current "fructose glut" in our food supply. We must learn from our experiences with ethanol and nicotine that regulation of the food industry, along with in-dividual and societal education, will be necessary to combat this fructose epidemic.
Article
Recent studies demonstrated Nϵ-(carboxymethyl) lysine (CML) in several tissue proteins. Incubation of proteins with glucose leads through a Schiff base to Amadori products. Oxidative cleavage of Amadori products is considered as a major route to CML formationin vivo,whereas it is not known which reactive oxygen species (ROS) is involved. The present study is undertaken to identify such a ROS. We prepared heavily glycated human serum albumin (HSA) which contained a high level of Amadori products, but an undetectable level of CML. Incubation of glycated HSA with FeCl2, but not with H2O2, led to CML formation which was enhanced by H2O2, but inhibited by catalase or mannitol, whereas superoxide dismutase had no effect. Similar data were obtained by experiments using Boc-fructose-lysine as a model Amadori compound. These data indicate that hydroxyl radical generated by the reaction of Fe2+with H2O2mediates CML formation from Amadori compounds.
Article
Insulin resistance induces nonalcoholic fatty liver disease and nonalcoholic steatohepatitis (NASH). We used a high-fat, high-calorie solid diet (HFD) to create a model of insulin resistance and NASH in nongenetically modified rats and to study the relationship between visceral adipose tissue and liver. Obesity and insulin resistance occurred in HFD rats, accompanied by a progressive increase in visceral adipose tissue tumor necrosis factor (TNF)-α mRNA and in circulating free fatty acids. HFD also decreased adiponectin mRNA and peroxisome proliferator-activated receptor (PPAR)-α expression in the visceral adipose tissue and the liver, respectively, and induced hepatic insulin resistance through TNF-α-mediated c-Jun N-terminal kinase (JNK)-dependent insulin receptor substrate-1Ser307 phosphorylation. These modifications lead to hepatic steatosis accompanied by oxidative stress phenomena, necroinflammation, and hepatocyte apoptosis at 4 weeks and by pericentral fibrosis at 6 months. Supplementation of n-3 polyunsaturated fatty acid, a PPARα ligand, to HFD-treated animals restored hepatic adiponectin and PPARα expression, reduced TNF-α hepatic levels, and ameliorated fatty liver and the degree of liver injury. Thus, our model mimics the most common features of NASH in humans and provides an ideal tool to study the role of individual pathogenetic events (as for PPARα down-regulation) and to define any future experimental therapy, such as n-3 polyunsaturated fatty acid, which ameliorated the degree of liver injury.