Download Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

G protein–coupled receptor wikipedia , lookup

Signal transduction wikipedia , lookup

Paracrine signalling wikipedia , lookup

Myokine wikipedia , lookup

Butyric acid wikipedia , lookup

Glycolysis wikipedia , lookup

Clinical neurochemistry wikipedia , lookup

Biochemistry wikipedia , lookup

Lipid signaling wikipedia , lookup

Basal metabolic rate wikipedia , lookup

Endocannabinoid system wikipedia , lookup

Specialized pro-resolving mediators wikipedia , lookup

Metabolism wikipedia , lookup

Hepoxilin wikipedia , lookup

Wilson's disease wikipedia , lookup

Fatty acid synthesis wikipedia , lookup

Ketosis wikipedia , lookup

Fatty acid metabolism wikipedia , lookup

Glyceroneogenesis wikipedia , lookup

Transcript
250
Fatty Acid and Glucose Sensors in Hepatic Lipid
Metabolism: Implications in NAFLD
Michele Vacca, MD, PhD1,2 Michael Allison, MD, PhD3
Antonio Vidal-Puig, MD, PhD2
1 Medical Research Council - Human Nutrition Research (MRC-HNR),
Cambridge, United Kingdom
2 Department of Clinical Biochemistry and University of Cambridge
Metabolic Research Laboratories, Institute of Metabolic Science,
Cambridge, United Kingdom
3 Liver Unit, Department of Medicine, Cambridge Biomedical Research
Centre, Cambridge University NHS Foundation Trust, United
Kingdom
4 Department of Biochemistry, University of Cambridge, United
Kingdom
Julian L. Griffin, PhD1,4
Address for correspondence Antonio Vidal-Puig, MD, PhD, FRCP,
FmedSci, TVPLab, Metabolic Research Laboratories Level 4, Institute of
Metabolic Science, Box 289, Addenbrooke’s Hospital, Cambridge CB2
0QQ, United Kingdom (e-mail: [email protected]).
Michele Vacca, MD, PhD, Medical Research Council – Human Nutrition
Research (MRC-HNR) & Institute of Metabolic Science, University of
Cambridge, Elsie Widdowson Laboratory, 120 Fulbourn Road,
Cambridge CB1 9NL, United Kingdom
(e-mail: [email protected]).
Semin Liver Dis 2015;35:250–261.
Abstract
Keywords
► insulin resistance
► mitochondrial
dysfunction
► peroxisome
proliferator-activated
receptors
► sterol regulatory
element binding
protein 1
► carbohydrate
response elementbinding protein
The term nonalcoholic fatty liver disease (NAFLD) covers a pathologic spectrum from
lipid accumulation alone (simple steatosis) to steatosis with associated inflammation
and fibrosis (nonalcoholic steatohepatitis [NASH]). Nonalcoholic steatohepatitis can
progress to cirrhosis and potentially to hepatocellular carcinoma. Although a genetic
predisposition has been highlighted, NAFLD is strongly associated with an unhealthy
lifestyle and hypercaloric diet in the context of obesity and metabolic disease. The
dysregulation of specific pathways (insulin signaling, mitochondrial function, fatty acid,
and lipoprotein metabolism) have been linked to steatosis, but elucidating the
molecular events determining evolution of the disease still requires further research
before it can be translated into specific personalized interventional strategies. In this
review, the authors focus on the early events of the pathophysiology of NASH, dissecting
the metabolic and nutritional pathways involving fatty acids and glucose sensors that
can modulate lipid accumulation in the liver, but also condition the progression to
cirrhosis and hepatocellular carcinoma.
The liver is a key player in the modulation of whole body
energy homeostasis, and hepatic dysfunction is usually associated with systemic metabolic unbalances and vice versa.
The nonalcoholic fatty liver disease (NAFLD) spectrum includes histologic changes ranging from simple steatosis to
steatohepatitis (NASH), fibrosis, and cirrhosis (developing
in 29% of NASH patients) ultimately leading (in 27% of
NASH-cirrhosis patients) to hepatocellular carcinoma (HCC)
development.1–3 Steatosis is defined as an intrahepatic triglyceride (TG) deposition exceeding the 95th percentile of TG
content (> 55 mg/g of liver) for lean healthy individuals, or as
the presence of micro- or macrovesicles in > 5% of hepato-
Issue Theme Nonalcoholic Fatty Liver
Disease; Guest Editors, Christopher P.
Day, FMedSci, and Quentin M. Anstee,
BSc, MBBS, PhD, MRCP(UK)
cytes.2,3 Often benign and self-limited, steatosis can progress
when hepatocyte injury (ballooning and cell death), inflammatory infiltrates and/or extracellular matrix deposition
develop, histologic changes diagnosed as nonalcoholic steatohepatitis (NASH). Nonalcoholic steatohepatitis has
emerged as the most rapidly increasing cause of chronic liver
disease; it is related to the epidemic of obesity extending to
both adults and children.4 Epidemiologic data has shown that
NAFLD is the typical hepatic manifestation of metabolic
syndrome (MetS), with a 50% prevalence of NAFLD in subjects
with diabetes, 76% in those with obesity, and 100% in those
morbidly obese with type 2 diabetes.3 Conversely, 50 to 100%
Copyright © 2015 by Thieme Medical
Publishers, Inc., 333 Seventh Avenue,
New York, NY 10001, USA.
Tel: +1(212) 584-4662.
DOI http://dx.doi.org/
10.1055/s-0035-1562945.
ISSN 0272-8087.
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
of subjects with NASH are overweight, 50 to 60% have
systemic hypertension, and 50 to 60% have dyslipidemia.3
When compared with the general population, NAFLD patients
also show an increased mortality and risk for cardiovascular
disease (CVD).1
The first step in the pathophysiology of NAFLD is the
accumulation of lipids in the liver that follow an imbalance
between lipid acquisition and removal. Whether steatosis can
progress toward NASH, or these entities represent two different diseases, is still highly debated. In fact, it can be argued
that when lipids are qualitatively inert, they lead to “simple
steatosis.” Alternatively, when they are reactive, they promote
lipotoxicity and NASH due to the formation of TG-derived
toxic metabolites (e.g., diacylglycerols, ceramides, cholesterol, reactive oxygen species) ultimately causing hepatocyte
dysfunction, lipoapoptosis, and the development of proinflammatory and fibrogenic stimuli.2 Sources of increased
lipid (TG) in the liver include excess dietary TG intake,
enhanced intrahepatic TG synthesis (fatty acids formed
from de novo lipogenesis [DNL]), excess fatty acid (FA) influx
into the liver from adipose tissue lipolysis, diminished export
of lipids from the liver, and reduced fatty acid oxidation (FAO).
Nonalcoholic fatty liver disease thus results from excessive
storage of lipid species occasionally exceeding the capability
of the hepatocytes to accommodate them physiologically.5 All
these biochemical events are under the control of a complex
system of signaling/transcriptional pathways, governed by
hormones, cytokines, adipokines, and nutrient-sensing transcription factors. In this review, we will focus on the events
that coordinately induce lipotoxicity, namely: (1) adipose
tissue dysfunction, lipotoxicity, and insulin resistance; (2)
reduced oxidative potential of the liver; (3) unbalances in
lipoprotein trafficking; (4) prolonged positive energy balance
(►Fig. 1); and on how they are controlled by insulin and
nutrient-sensing transcription factors.
Adipose Tissue Dysfunction, Lipotoxicity,
and Insulin-Resistance
Patients with NAFLD typically present with hyperinsulinemia
and insulin resistance (IR). Insulin resistance originates from
the convergence of genetic predisposition and environmental
stressor factors, and has strong negative effects in liver
metabolism.2,6 In the liver, insulin is essential to control
the “fast-to-fed” metabolic shift in hepatic carbohydrate
metabolism (promoting hepatic glucose uptake, glycolysis,
and glycogen synthesis; inhibiting gluconeogenesis and glycogen utilization), and to modulate lipid metabolism (inhibiting FAO; promoteing DNL, free fatty acid [FFA] influx, and
very low-density lipoprotein [VLDL] secretion) thus leading
to hepatic lipid accumulation and to the promotion of hepatic
triglyceride export (inducing hypertriglyceridemia6,7). Under
physiologic conditions, insulin also inhibits lipolysis and FFA
release from adipose tissue (AT) repressing the activity of
hormone-sensitive lipase (HSL) (►Fig. 2).6,7 As a consequence
of hyperinsulinemia (with/without hyperglycemia) excess
glucose in the liver is converted to glycogen or is used to
produce acetyl-CoA (via glycolysis) to be either further
Vacca et al.
Fig. 1 Nutrient fate in relationship to caloric intake. During isocaloric
balanced feeding, nutrients are mainly used to produce energy and for
structural purposes (e.g., membranes, proteins, organelles, DNA).
Only a small quantity of these nutrients is stored during the fed states
(to be released during fasting). A continuous hypercaloric feeding is
associated with the need of storing excess calories. Cells have limited
carbohydrate-storage capacity as glycogen; as a consequence, proteins and carbohydrates are converted into fatty acids and triglycerides having neither osmotic nor colloidal effects for the cell, and
providing high caloric density (9 kcal/g).TGs, triglycerides.
metabolized or, alternatively, utilized as a substrate for
DNL.6,7 De novo lipogenesis facilitates the synthesis of saturated fatty acids (SFAs) for TG formation7 that can be stored
within hepatocytes or, alternatively, packaged into VLDL
lipoproteins to be secreted into the circulation (to be subsequently used in peripheral organs or stored in adipocytes).2
Triglycerides provide high caloric density (9 kcal/g), are
insoluble in water, and have neither osmotic nor colloidal
effects for the cell2; thus DNL is the preferred mechanism to
stock excess energy within the body. When DNL is overactivated in the liver because of hyperinsulinemia, it promotes hepatic fat accumulation leading to NAFLD, and
hypertriglyceridemia. Adipose tissue function is thus an
important factor in the pathophysiology of NAFLD and its
progression toward NASH as shown by the fact that loss of
function of key genes in adipogenesis promotes steatosis and
the progression of liver damage,8–10 and variants in genes
known to be associated to steatosis (e.g., PNPLA3) also affect
AT distribution.11 In health, AT protects the body from an
excessive exposure to fatty acids; when adipocyte expandability is exhausted, this event is associated with lipotoxicity
(the accumulation of toxic fatty acids derivatives in organs
different from AT) and peripheral insulin resistance. Adipose
tissue acquires a proinflammatory profile (suppression of
adiponectin, promotion of adipocytokines) and these events
in the liver lead to accumulation of lipid toxic species, reactive
oxygen species (ROS) production, endoplasmic reticulum
stress, activation of stellate cells, and progression of liver
damage.12,13 Adipose tissue dysfunction and IR are early
processes in the development of hepatic damage: In a population of “healthy” (nonobese, nondiabetic, normolipidemic)
biopsy-proven NAFLD patients, hyperinsulinemia, and
Seminars in Liver Disease
Vol. 35
No. 3/2015
251
252
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
Vacca et al.
Fig. 2 Role of hepatic fatty acid metabolism in the development of hepatic steatosis. Hepatic lipid accumulation can result from (1) a prolonged positive
energy balance, (2) adipose tissue dysfunction and insulin resistance, (3) impairment of fatty acid oxidation and mitochondrial function, and (4) unbalances
in lipoprotein trafficking. Insulin-mediated activation of SREBP1 proteins and glucose-mediated activation of ChREBP play a major role in these processes.
Continuous arrows represent direct biochemical reactions, dash arrows represent modulatory mechanisms. ApoEr, ApoE receptor; FAT/CD36, fatty acid
translocase/cluster differentiation protein-36; CETP, cholesteryl ester transfer protein; ChREBP, carbohydrate response element-binding protein; CM,
chylomicrons; HDL, high-density lipoprotein; HSL, hormone-sensitive lipase; IDL, intermediate-density lipoprotein; LDL, low-density lipoprotein; LDLr, lowdensity lipoprotein receptor; OxLDL, oxidized low-density lipoprotein; Ox-Phos, oxidative phosphorylation; sLDL, small low-density lipoprotein; SREBPs, sterol
regulatory element binding proteins; TG, triglyceride; VLDL, very low-density lipoprotein.
AT/hepatic IR already coexist at the time of the diagnosis and
progressively increase across NAFLD stages; of note, AT IR
independently predicts liver histology and alters cardiometabolic risk parameters.14 Conversely, NAFLD can occur also in
the absence of peripheral IR.3,5 Rats fed with a short-term
high fat diet (HFD) develop hepatic fat accumulation and
hepatic IR in the absence of changes in the whole body IR.15
Even if it is still debated whether peripheral or hepatic IR
arises first, adipocyte dysfunction represents a contributing
factor to the onset of lipotoxicity and (both peripheral and
hepatic) IR, and could have a major role for the progression of
fatty liver disease.
Fatty Acid Oxidation, Mitochondrial
Function, and the Progression of NASH
Fatty acid β-oxidation allows the oxidation of FAs into acetylCoA, a substrate for oxidative phosphorylation (OXPHOS),
ketone body formation, or for other biochemical reactions
(e.g., gluconeogenesis or DNL).16,17 In normal conditions,
β-oxidation occurs in mitochondria more than in peroxisomes. Mitochondrial β-oxidation is mostly controlled by
Seminars in Liver Disease
Vol. 35
No. 3/2015
the uptake of FA into the mitochondrial matrix via carnitine-palmityl-transferase 1 (CPT-1), which is inhibited by
malonyl-CoA.16,17 Fatty acids can be metabolized exclusively
in the presence of oxygen as the final acceptor of the reducing
equivalents deriving from their oxidation by the four mitochondrial respiratory complexes. The mitochondrial respiratory complex couples the electron transfer activities to Hþtranslocation from the matrix to the outer side of the mitochondrial inner membrane thus building up a transmembrane electrochemical potential (Δψ).16,17 When adenosine
diphosphate (ADP) is available, this electrochemical gradient
is utilized for adenosine triphosphate (ATP) synthesis by ATP
synthase, or it can otherwise exert a negative control feedback on the respiratory flux.16,17 Furthermore, ROS overproduction occurs when mitochondrial FAO supplies excessive
respiratory substrates in the absence of a concomitant upregulation of mitochondrial respiratory chain activity, increases
mitochondrial membrane potential (Δψ), or produces imbalances in the ATP/ADP ratio causing stalling of the respiratory
flux.16,17 Excess FAs and ROS also promote uncoupling proteins (UCPs) activation that lower the mitochondrial Δψ and
the intracellular oxygen tension increasing the respiration
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
rate (thus leading to reduced ROS production); this is a relay
system that, when overactivated (e.g., in obesity and steatosis), affects OXPHOS efficiency.16 Thus, FAO itself promotes
the saturation of the oxidative system in steatosis, and leads
to mitochondrial dysfunction and to the promotion of the
extramitochondrial (microsomal and peroxisomal oxidation)
oxidation systems.16–18 Ω-oxidation of FAs is normally a
minor pathway of FAO because one of its key enzymes,
cytochrome P450 4A (CYP4A), is inhibited by insulin. Hence,
IR leads to a significant overactivation of this pathway that
produces long-chain dicarboxylic acids that further inhibit
mitochondrial function.16–18 Long-chain dicarboxylic acids
(and very long chain FAs) can be metabolized by the peroxisomal β-oxidation to complete their oxidation processes in
mitochondria; the genetic ablation of enzymes involved in
the peroxisomal β-oxidation leads to hepatic steatosis.19
Mitochondrial dysfunction and the activation of the extramitochondrial oxidation systems are thus key events in the
development of a lethal vicious cycle able to generate a
dramatic amplification of ROS production and liver damage
that promotes the progression of NAFLD into NASH. The
mitochondria are also the primary targets ROS-induced damage: Oxidative modifications of the respiratory complexes are
associated with the impairment of their catalytic functions,
mutagenesis of the mitochondrial DNA, mitochondrial dysfunction, and propagation of the oxidative damage, thus
leading to hepatocellular death and NASH progression.17
Strategies aimed to improve the cellular REDOX homeostasis
by supplying the hepatocyte with nonenzymatic antioxidant
molecules (such as vitamin E20) could thus be a promising
approach to the management of NASH.
If Lipoproteins Deal with Liver’s Fat, What
Goes Wrong in Steatosis?
The liver is the major regulator of metabolite flux in the body,
processing lipids provided by diet and AT-released FFAs
(contributing in equal amounts), and to a smaller part by
DNL in the liver itself. During the development of IR, the
massive increase of FFA and the promotion of DNL render
lipoprotein trafficking efficiency a critical step in the pathophysiology of NAFLD.2,21 The fate of newly formed or dietary
lipids is to be oxidized, or to be esterified to TGs, incorporated
into VLDL, secreted into the systemic circulation and delivered to the peripheral tissues. Very low-density lipoprotein
particles are constituted by apolipoprotein (Apo) B-100
(ApoB-100) and C (ApoC), lipids (TGs and cholesterol esters),
and phospholipids. Within the hepatocytes, microsomal
transfer protein (MTP) promotes ApoB-100 fusion with TGs
droplets, and VLDL formation and secretion.2,21 In the initial
phases of steatosis, lipid accumulation is counterbalanced by
increased VLDL secretion, but this adaptive mechanism
reaches a point at which it is inadequate to compensate for
the degree of intracellular TG accumulation.22,23 In fact, the
VLDL-ApoB-100 secretion rates (the rate-limiting step in
VLDL formation) is not increased in subjects with steatosis,
thus leading to the production of very large TG-rich VLDL
(that cannot be secreted through the sinusoidal endothelial
Vacca et al.
pores) and impairment of VLDL synthesis and export.24
Subsequently, serum levels of LDL and of the atherogenic
small LDL (sLDL) also increase due to the promotion of
cholesteryl ester transfer protein (CETP; enriches VLDL in
cholesterol), and of hepatic lipase ([HL] regulates plasma
concentration of the atherogenic lipoproteins intermediatedensity lipoproteins -IDL- and of sLDL). Both CETP and HL
activities are increased in NAFLD and may favor the formation
of sLDL.2
Dietary lipids are absorbed by enterocytes, incorporated in
TG-rich chylomicrons, transported into the systemic circulation, and then delivered to the liver (20%) or peripheral tissue
(80%), where lipoprotein lipase ([LPL] the rate-limiting step of
TG uptake) releases FFAs for uptake.2 One of the strongest
inhibitors of LPL is apolipoprotein C-III (ApoC-III; an essential
constituent of VLDL particles), and a loss of function of this
apolipoprotein has been associated with obesity and metabolic complications due to an enhanced peripheral activity of
LPL.25 On the other hand, the overexpression of ApoC-III
promotes liver steatosis and hepatic insulin resistance due
to reduced peripheral FFA uptake, increased hepatic triglyceride delivery, and surprisingly, a reduction in hepatic triglyceride secretion.26 Interestingly, insulin and glucose exert
opposite effects on ApoC-III, which is promoted by glucose
and inhibited by insulin27,28; insulin instead promotes the
expression of transporter fatty acid translocase / cluster
differentiation protein-36 (FAT/CD36 mediates the uptake
and the intracellular transport of FAs), thus facilitating lipid
accumulation in the liver.29 In individuals with IR, insulin fails
to suppress efficiently ApoC-III in the liver (thus leading to LPL
inhibition in peripheral tissues, and to the increase of the
hepatic uptake of TGs-rich chylomicrons remnants) and
hormone-sensitive lipase (HSL) in AT (thus leading to an
increase of adipose-derived FFA flux to the liver).29,30 Hepatic
IR thus promotes a progressive deterioration of steatosis and a
deleterious lipoprotein profile; this is the strongest evidence
linking hepatic steatosis to the consequences of obesity and
metabolic syndrome.
Diet, Nutrient Sensors, and Modulation of
Lipid Metabolism
Sugar or Fat: Which Is the Enemy?
Insights for addressing the role of excess calories and diet
composition in the pathophysiology of NAFLD come from the
study of animal models fed high-energy diets. Although not
standardized in macronutrient content or composition, these
studies show that prolonged energy excess, especially in the
form of high carbohydrate or fat, rapidly induces fatty liver
with differences in the progression of liver injury potentially
depending on macronutrient composition.31,32 When energy
balance is positive, mammalian livers preferentially use glycolysis to generate energy, while excess glucose is stored in
the form of glycogen (glycogen synthesis) and fatty acids
(DNL) that will be stored in the adipose tissue in the form of
triglycerides; when positive excess energy intake is prolonged, the liver is also able to store significant quantities
of lipids leading to hepatic steatosis (►Fig. 1).31,32 In terms of
Seminars in Liver Disease
Vol. 35
No. 3/2015
253
254
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
macronutrient composition, high sucrose/fructose diets induce intrahepatic lipid accumulation independently from
their fat content, and do not necessarily progress toward
steatohepatitis.32–34 On the other hand, it has also been
shown that “high-fat” diets (35–70% of total calories constituted by fat) induce steatosis and liver damage not only due to
fat content, but also due to their content in simple sugars. For
example, ketogenic diets (very high in fat with almost no
carbohydrate) neither induce massive steatosis nor raise
alanine aminotransferase (ALT) outside the normal range.35
Saturated fat is able to stimulate both FAO via a peroxisome
proliferator activated receptor-α (PPAR-α) dependent mechanism and DNL in the liver, but the prominence of these
effects seems to depend on sucrose content;36,37 when
saturated fat is consumed without sucrose, FAO promotion
is the prominent feature, thus leading to minimal hepatic
steatosis or liver injury.37 Diets that incorporate both macronutrients (carbohydrate and fat) thus appear to represent a
situation closer to the so-called “Western” diet; such diets
(enriched in saturated fat, cholesterol, sucrose and/or fructose) induce obesity, insulin resistance, and hepatic steatosis,
along with different degrees of hepatic injury, inflammation,
and mild fibrosis.38–40 The types of FA have also been invoked
as a factor contributing to differences in the capability of diets
to induce steatosis, for example, both small/medium-chain
FAs (that need more energy to be utilized) and long-chain
saturated FAs alone (stimulating FAO by promoting PPARα
and its coactivator PGC-1-α) fail to induce massive steatosis,36,37,41 whereas transfats (> 15% of total calories)42,43
promote hepatic steatosis even in the absence of free sucrose.
The role of polyunsaturated fatty acids (PUFAs) is still discussed because they decrease liver triglyceride content and
plasma ALT levels in leptin-deficient mice,44 but when administered in excess (> 35% of total calories) via total enteral
nutrition, PUFAs overfeeding causes steatosis and liver
damage.45
Therefore, micronutrient composition could affect the
capability of a specific diet to induce DNL and/or FAO. Further
studies are necessary to assess whether specific nutrient
mixtures cause liver injury in excess to their ability to induce
hepatic steatosis.
Sterol Regulatory Element Binding Protein 1c and
Carbohydrate-Response Element-Binding Protein:
Partners in DNL Promotion
De novo lipogenesis is finely regulated at the transcriptional
level mainly by two transcription factors, the sterol regulatory element binding protein 1 (SREBP1), and the glucose
sensor carbohydrate response element-binding protein
(ChREBP), which are activated by insulin and glucose, respectively. Both these transcription factors promote the expression of key enzymes for DNL (e.g., ATP citrate lyase [ACL];
acetyl-CoA carboxylase [ACC]; fatty acid synthase [FAS]).7,46
Insulin, SFAs, and the oxysterol sensor liver X receptor
(LXR) promote SREBP1c,47,48 whereas PUFAs, glucagon, and 5'
AMP-activated protein kinase (AMPK) signaling inhibit
it.44,48,49 The relationship between insulin and SREBP-1c is
finely regulated: Insulin promotes the expression of a SREBPSeminars in Liver Disease
Vol. 35
No. 3/2015
Vacca et al.
1-mediated negative loop that activates DNL, but limits an
excessive activation of this metabolic pathway toward the
promotion of SREBP-1c inhibitor insulin-induced gene 1
(Insig1)50,51; SREBP-1c also negatively feedbacks insulin signaling through the inhibition of insulin receptor substrate 2
(IRS2), thus silencing phosphatidylinositol 3-kinases/protein
kinase B, PI3K/Akt cascade.52,53 In obesity, hyperinsulinemia
thus boosts SREBP-1c activation, which promotes DNL, negatively feedbacks insulin signaling (leading to decreased glycogen synthesis and increased gluconeogenesis), and directly
promotes gluconeogenesis (e.g., phosphoenolpyruvate
carboxykinase [PEPCK]), thus inducing steatosis and
hyperglycemia.7,52
Increased intracellular glucose levels activate ChREBP54
resulting in the induction of the transcription of genes
involved in glycolysis (e.g., liver pyruvate kinase [LPK]) and
DNL,55 causing a further worsening of steatosis.5 In murine
models of obesity, liver-specific adenovirus-mediated inhibition of ChREBP markedly impairs lipogenic gene expression in
the liver, protects from hepatic steatosis, ameliorates insulin
signaling and overall glucose tolerance, and also decreases
plasma levels of triglycerides and nonesterified fatty acids.56
In patients with NASH, ChREBP expression correlates with the
degree of steatosis; however, its expression decreases in the
presence of severe insulin resistance.57 ChREBP overexpression promotes DNL genes and stearoyl-CoA desaturase-1
(Scd1), a critical regulator of energy metabolism that catalyzes the transformation of SFAs into monounsaturated fatty
acids (MUFAs). As a consequence, ChREBP activation results in
a prevalent accumulation of MUFAs that does not impact
much on insulin sensitivity.57 In this the concomitant knockdown of Scd1 worsens insulin sensitivity.57 These findings are
in line with a previous report showing that liver-specific Scd1
knockout (KO) mice are protected from steatosis when fed a
high-carbohydrate very low-fat diet.32
Therefore, SREBP1c and ChREBP activation in hyperinsulinemic subjects has a crucial role in the development of
hepatic lipid accumulation (►Fig. 2). The relative balance
between SREBP1c and ChREBP contribution in modulating
the lipogenic pathway could have different outcomes for the
accumulation of specific lipid species and for the degree of IR,
and could potentially explain different outcomes in terms of
MetS complications.58,59
Peroxisome Proliferator-Activated Receptors and Their
Coactivators in the Modulation of Hepatic Lipid
Accumulation
Lipid-sensing nuclear receptors (NRs) are transcription factors participating in the control of complex processes in living
organisms. Nuclear receptors show considerable specificity in
their activation and tissue-specific expression.60 In the human genome, 48 NRs have been identified, while in rodents
there are 49.61 A subfamily of these transcription factors,
known as “adopted-orphan NRs” acts as lipid sensors and
have been involved in the pathophysiology of MetS.62 Of
particular interest for the pathophysiology of NAFLD, are
the “fatty acid sensors” peroxisome proliferator-activated
receptors (PPARs) and their coactivators peroxisome
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
proliferator-activated receptor γ coactivators 1 (PGC1s;
see ►Table 1). Peroxisome proliferator-activated receptors
(PPARα/NR1C1; PPARβ/δ/NR1C2; PPARγ/NR1C3) form heterodimers with the retinoid X receptors (RXR) to control
metabolic programs and to regulate energy homeostasis.62
Peroxisome proliferator-activated receptors bind to specific
DNA consensus sites termed PPAR response elements
(PPREs). Although sharing sequence and structure homology,
PPARs are characterized by specific ligand-binding domains
(that can accommodate a wide diversity of natural and
synthetic compounds), tissue distributions, and physiologic
functions.63
Peroxisome Proliferator-Activated Receptor α
Peroxisome proliferator-activated receptor α (PPARα) is
prominently expressed in the liver (also expressed at a lower
level in kidney, heart, skeletal muscle, small intestine, brown
AT, and immune cells) and can be activated by fatty acids, 1palmitoyl-2-oleoyl-sn-glycerol-3-phosphocholine, fibrates,
highly selective PPARα agonists (e.g., LY518674), and
SPPARMs (selective PPAR modulators).63 The major physiologic function of PPARα is to promote fatty acid utilization
during fasting to allow gluconeogenesis,44,64,65 but PPARα is
also one of the key regulators of the response to excess fat. In
fact, PPARα modulates fatty acid uptake (fatty acid binding
protein [FATP], CD36, CPT-1) and lipid trafficking (promotion
of LPL and apoA proteins; inhibition of the LPL inhibitor
apoCIII and of proprotein convertase subtilisin/type 9
[PCSK9]), promotes β-oxidation (acyl-CoA oxidase [ACOX],
thiolase, acyl-CoA-dehydrogenase, cytochrome P450-ω-hydroxylase), and inhibits DNL.64,66–69 Given the ability of
PPARα agonists to modulate both TG and HDL metabolism,70,71 fibrates are thus employed for the management
of primary hypertriglyceridemia and mixed hyperlipid-
Vacca et al.
emia.72,73 Peroxisome proliferator-activated receptor α has
also shown a potential in the regulation of the pathophysiologic events causing liver steatosis. Following a high-fat diet
(HFD), high fructose or methionine-choline deficient ([MCD];
a model to induce NASH) diets, PPARα null mice develop
hypertriglyceridemia and are more susceptible to steatosis
and to the progression of NASH due to an impairment of
mitochondrial fatty acid β-oxidation, and to increased hepatic
oxidative stress and expression of proinflammatory genes.
These mice also develop massive steatosis, lobular inflammation, with a significant increase of the NASH activity score
when compared with wild-type animals.74–76 Conversely,
PPARα ligands protect the liver from NAFLD and NASH,
promoting fatty acid oxidation, and reducing endoplasmic
reticulum stress, hepatic insulin resistance, glucose intolerance, and levels of expression of inflammatory genes.74,77–79
Despite the robustness of this evidence in animal models,
only few trials with relatively low numbers of patients have
been performed to confirm these results in humans, and have
not so far provided additional support for significant efficacy
in NASH. Although clofibrate was not effective in the management of NASH,80 fenofibrate was able to ameliorate the
degree of metabolic impairment (especially fasting glucose,
triglycerides, and apolipoprotein A1), and improve liver
enzymes in a small cohort of patients with NASH; however,
its effects on liver histology were minimal.81
Peroxisome Proliferator-Activated Receptor β/δ
Peroxisome proliferator-activated receptor β/δ (PPARβ/δ)
exhibits a broad expression pattern63,82 and can be activated
by polyunsaturated fatty acids (eicosapentaenoic acid [EPA],
arachidonic acid [ARA], and dihomo-γ-linoleic acid), prostaglandins (PGs; PGA1, PGE2, PGD2, prostacyclin -PGI- and its
stable analogue carba-prostacyclin), 4-hydroxynonenal, and
Table 1 Potential relevance of peroxisome proliferator-activated receptor (PPARs) and peroxisome proliferator-activated receptor γ
coactivators (PGCs) in the pathophysiology of nonalcoholic fatty liver disease
PPARα
PPARβ/δ
De novo lipogenesis
↓
?
Fatty acid oxidation & mitochondrial function
"
"
Fatty acids uptake
"
/
Lipoprotein transport
"
"
Gluconeogenesis
"
Glycolysis
PPARγ
PGC1α
PGC1β
/
"
"
"
"
"
"
/
"
"
"
/
" (hepatic)
↓ (AT/systemic)
"
/
"
"
"
"
"
"
Inflammation/damage
↓
↓
↓
↓
↓
Steatosis
↓
?
" (hepatic)
↓ (AT/systemic)
↓
↓
Hepatic insulin sensitivity
"
"
↓ (hepatic)
" (AT/systemic)
"
¼
Triglycerides
↓
↓
" (hepatic)
↓ (AT/systemic)
↓
"
Abbreviations: ?, contradictory results; /, not investigated; ", promoted; ↓, inhibited; ¼, unchanged; hepatic, effects of hepatic-specific modulation;
AT/systemic, effects of systemic or adipose tissue-specific modulation.
Seminars in Liver Disease
Vol. 35
No. 3/2015
255
256
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
selective synthetic agonists (GW501516, GW0742).83–85 In
the liver, PPARβ/δ controls fatty acid oxidation and the
uncoupling of oxidative phosphorylation through activation
of its target genes such as long chain acyl-CoA dehydrogenase
(LCAD), CPT-1, ACOX1, long chain acyl-CoA-synthetase (ACSL)
and UCP-1. Depending on the organ targeted and the experimental design used, PPARβ/δ has shown positive or negative
action on liver steatosis, but an overall beneficial action on its
progression. Adipose tissue-specific overexpression of
PPARβ/δ in rodents protects from diet-induced obesity, hypertriglyceridemia, and hepatic steatosis.86 In models of
obesity-induced steatosis (HFD and ob/ob), pharmacologic
or liver-specific adenoviral activation of PPARβ/δ promotes
hepatic fatty acid oxidation (increase of plasma β-hydroxybutyrate and of mRNA expression of PPARα and its target
genes, promotion of the AMPK/PGC-1α-lipin1/PPARα pathway) and the Krebs cycle,87,88 but also stimulates (AMPKmediated) insulin sensitivity, thus reducing fasting glucose
levels, promoting hepatic glycogen deposition, and glycolysis.87,89 The effects of PPARβ/δ activation on DNL are controversial because some authors show that PPARβ/δ directly
promotes the transcription of Insig-1 (thus leading to
SREBP1c inhibition, DNL suppression, and reduced hepatic
steatosis in obese diabetic db/db mice), whereas other authors indicate that PPARβ/δ induces hepatic lipid production
mainly in the form of unsaturated (rather than saturated)
fat.89,90 In both cases, liver damage has been shown to be
reduced,89,90 and this goes in line with the inhibitory effect of
PPARβ/δ on STAT3 phosphorylation that prevents the activation of proinflammatory signals (e.g., tumor growth factor β1,
interleukin- [IL-] 6, IL-1β, MCP-1, tumor necrosis factor α,
nuclear factor-κB1).91,92 Animal and human studies also
provide evidence that PPARβ/δ is beneficial on lipoprotein
metabolism because PPARβ/δ activation increases apoAI,
apoAII, and HDL-cholesterol, while reducing small dense
LDL levels.93–97 Apparent discrepancies in the role of
PPARβ/δ to modulate DNL could be thus explained by the
different experimental designs (i.e., obesity models, PPARβ/δ
activation strategies) that preferentially select for specific
functions of PPARβ/δ, depending on the pathophysiologic
context. It seems likely that in the liver PPARβ/δ physiologically reacts to an excessive accumulation of lipids and/or
products of oxidative stress and/or proinflammatory mediators (prostaglandins) to limit further accumulation of fat (by
promoting FAO), to prevent hepatic insulin resistance (by
promoting AMPK/PGC1α axis), and to limit further FA production associated with an excessive diet-induced insulinmediated activation of DNL (by promoting Insig-1). Nevertheless, analysis of the PPARβ/δ function in the progression of
NASH deserves further attention because it has been shown
that it promotes stellate cell activation and fibrosis (upon
CCL4 challenge),98 and its role in hepatic proliferation and
hepatocarcinogenesis is contentious.99,100
Peroxisome Proliferator-Activated Receptor γ
The PPARγ gene encodes for three splicing variants giving rise
to two functional proteins (PPARγ1 and PPARγ3 transcripts
encode for PPARγ1 and is expressed in AT, immune cells,
Seminars in Liver Disease
Vol. 35
No. 3/2015
Vacca et al.
intestine, liver, and kidney; PPARγ2 transcript encodes for a
protein expressed in ATs). Peroxisome proliferator-activated
receptor γ is an important transcriptional regulator of adipocyte differentiation and function.9,10,63 Mutations in the
PPARγ gene (e.g., familial partial lipodystrophy type 3
[FPLD3], PPARγ Pro12Ala, C1431T polymorphisms) induce
severe insulin resistance, dyslipidemia, steatosis, and earlyonset hypertension.101–105 A wide spectrum of PPARγ ligands
has been identified, ranging from the potential natural
ligands including monounsaturated and polyunsaturated
FA, eicosanoid FA-derivatives (hydroxyoctadecanoic acid
[HODE]; 13-HODE and 9-HODE), and prostanoids (e.g.,
15-deoxy-Δ12,14-prostaglandin J2) to the pharmacologic agonists thiazolidinediones (TZDs), selective PPARγ modulators
(SPARMs), partial PPARγ/α agonists (e.g., TDZ18), curcumin,
protopanaxatriol, and harmine.72,106 Thiazolidinediones
(pioglitazone and rosiglitazone) have been approved for
clinical use in type 2 diabetes, and proposed as possibly
effective in NASH.107 The primary therapeutic effect of
TZDs is to promote the expansion of subcutaneous AT due
to an enhanced differentiation of preadipocytes in mature
adipocytes. This event promotes lipid repartitioning from
visceral fat, liver, and skeletal muscle to the subcutaneous AT,
preventing lipotoxicity-induced insulin resistance and increased circulating levels of FFA.8,9,108 Peroxisome proliferator-activated receptor γ also stimulates adiponectin
secretion (by promoting AMPK signaling and FA oxidation,
while inhibiting hepatic gluconeogenesis)63,109 and is a negative modulator of the proinflammatory cascades in the liver
thus potentially preventing the progression of NASH.110
Tissue-specific PPARγ modulation strategies have provided
the evidence that PPARγ exerts its beneficial effects on liver
steatosis mainly due to its action in AT.8,10,111–115 A targeted
deletion of PPARγ in AT is associated with elevated plasma
levels of FFAs and triglycerides and increased susceptibility to
HFD-induced steatosis, hyperinsulinemia, and IR.8 On the
other hand, hepatocyte-specific PPARγ-knockout mice are
protected against HFD-induced hepatic steatosis, show improved glucose tolerance, and reduced gluconeogenesis. Peroxisome proliferator-activated receptor γ-knockout mice are
also characterized by a downregulation of genes involved in
lipogenesis (SCD1, SREBP-1c, and ACC), lipid transport (CD36/
FAT, L-FABP, and MTP), and β-oxidation (PPARα and ACO).115
As a confirmation of a candidate “pro-steatotic” action of
PPARγ into the liver, adenoviral PPARγ hepatic overexpression
causes hepatic steatosis,116 overexpression of genes involved
in fatty acid transport, and triglyceride synthesis.114,116
However, the pharmacologic systemic activation of PPARγ
using TZDs has been suggested to be potentially beneficial in
the management of NAFLD20 because the net result of systemic PPARγ activation is to (1) increase AT lipid uptake and
storage, (2) decrease serum FFA and TG, (3) increase peripheral glucose absorption, and (4) enhance energy expenditure.
Altogether these effects improve insulin sensitivity, and
reduce hepatic gluconeogenesis and steatosis.9,63 Unfortunately, although effective in controlling glycosylated hemoglobin and reducing liver steatosis, the enthusiasm for TZDs
as therapeutic agents has been limited in recent years
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
because they are not devoid of side effects such as weight gain
(2–3 kg per 1% glycosylated hemoglobin lowered), anemia,
pulmonary edema, congestive cardiac failure, and increased
risk of myocardial infarction.117 Thus rosiglitazone was voluntarily withdrawn from the market despite being one of the
most effective drugs in reducing glycosylated hemoglobin.
Peroxisome Proliferator-Activated Receptor γ
Coactivators
A more radical approach to the treatment and the prevention
of NASH could be the modulation of lipid homeostasis by
strategies aiming at inducing peroxisome proliferator-activated receptor γ coactivator 1 (PGC1) α and β. First characterized as important regulators of mitochondrial biogenesis
and function, PGC1s have been described as central coordinators of metabolic homeostasis and their modulation appears to be potentially beneficial in NAFLD.
Peroxisome proliferator-activated receptor γ coactivator
1α coactivates multiple transcription factors (FoxO1, glucocorticoid receptor, hepatic nuclear factor-4α, estrogen-related receptor-α, and PPARα), thus acting as a key promoter of
gluconeogenesis, fatty acid oxidation, lipid transport, and
oxidative phosphorylation during times of nutrient deprivation.118 Hepatic PGC1α transcription is induced by fasting
(because it is promoted by glucagon and inhibited by insulin),
HFD, obesity, and insulin resistance. PGC1a is also tightly
modulated at posttranscriptional level (e.g., by the AMPK/
SIRT pathway that is activated by cold, exercise, metformin,
resveratrol, and other stimuli).118 PGC1α KO mice develop
metabolic abnormalities in energy-demanding organs that
lead to increased body fat, susceptibility to cold and, following short term starvation, hepatic steatosis (due to a combination of reduced mitochondrial respiratory capacity,
promotion of lipogenic genes, and increased supply of
FAs).119 Hepatic-specific PGC1α heterozygous mice show
hypertriglyceridemia (due to increased expression of genes
involved in triglyceride-rich lipoprotein assembly), hepatic
steatosis and diminished ketogenesis during fasting (decreased expression of genes involved in β-oxidation). These
mice also develop hepatic insulin resistance following HFD.120
Interestingly, mice lacking SIRT1 (one of the upstream regulators of the PGC1α/PPARα pair) in the liver show a similar
phenotype. When fed with HFD, SIRT1 KO mice develop
hepatic steatosis, hepatic inflammation, and endoplasmic
reticulum stress due to the suppression of PGC-1α/PPARα.121
Altogether, these data indicate that pharmacologic modulation
of AMPK/SIRT1 cascade could be an effective approach to
preventing obesity-associated metabolic diseases.
Peroxisome proliferator-activated receptor γ coactivator
1β is considered an important regulator of metabolism in the
liver because it coactivates different key transcription factors
(including estrogen-related receptors, nuclear respiratory
factor 1, liver X receptors, etc.) involved in mitochondrial
biogenesis, oxidative metabolism, lipogenesis, and TG secretion. HFD stimulates not only the expression of SREBP1 in
liver, but also of PGC1β, which contributes to the induction of
the lipogenic program facilitating the release of triglycerides
and cholesterol in VLDL particles. However, unlike SREBP1,
Vacca et al.
PGC1β reduces fat accumulation in the liver through concomitant stimulation of FAO and lipoprotein transport.122 The
global knockout of PGC1β, although not showing overt phenotype under normal laboratory conditions, is characterized
by a general defect in the expression of genes involved in the
electron transport chain of the mitochondria; and, in response to acute high fat dietary loads, the lack of PGC1β
impairs hepatic lipid metabolism in the liver, resulting in
hepatic steatosis123,124 and reduced lipoprotein-associated
triglyceride and cholesterol content.123 Conversely, MCD-fed
hepatocyte-selective PGC1β transgenic mice are protected
from lipid overload and from the progression to fibrosis due
to a coordinated promotion of mitochondrial function and
lipoprotein metabolism, and a reduction of oxidative
stress.125
In the context of liver steatosis, the selective modulation of
PGC1 proteins and their targeted pathways is thus a candidate
strategy to protect the liver from lipid accumulation and
oxidative damage due to the pleiotropic action of these
coactivators in lipid homeostasis. However, to date the strategies aiming at activating this pathway rely on indirectly
modulating PGCs upstream cascades (e.g., with metformin,
resveratrol, etc.) or on stimulating the relevant coactivated
receptor (e.g., PPARs), whereas mechanisms to target PGC1
proteins directly in humans are still being sorted.
Conclusion
Hepatic steatosis and NASH are the hepatic manifestation of
lipid-induced toxicity in the context of MetS, and are closely
related to excessive dietary calorie intake, obesity, and
impaired metabolic homeostasis. The close interrelationship
between obesity, AT dysfunction, onset of IR, and NAFLD have
been widely characterized, with defective insulin signaling,
impaired mitochondrial function, and dysregulated lipoprotein trafficking being the key effectors leading to hepatic TG
accumulation. However, the exact natural history of the
disease and the factors responsible for the progression
toward liver damage still remain elusive. Although lipotoxicity, oxidative stress, exogenous factors (e.g., drugs, microbiota, alcohol, and fructose consumption), and genetic
predisposition have been proposed as modulators in this
progression, more detailed insights are needed to elucidate
the mechanisms responsible for the evolution of NAFLD
toward more severe forms of hepatic disease. Currently,
the modulation of lifestyle through diet and exercise and
the management of obesity, insulin resistance, and diabetes
are the toolkit of management with proven positive effects
on steatosis likely to reduce the risk of progression of
established NASH. However, a more detailed characterization of the pathways that define lipid-related reactive toxic
species that cause the lipid-related hepatocyte cell death
inflammation as well as those attempting to promote regeneration, hepatic stellate cell activation, and extracellular
matrix deposition may provide an integrated picture of the
pathophysiology of NASH and identify novel therapeutic
approaches to prevent its progression toward cirrhosis and
HCC.
Seminars in Liver Disease
Vol. 35
No. 3/2015
257
258
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
Vacca et al.
Abbreviations
Acknowledgments
Prof. Vidal-Puig was funded by the MRC and BHF program
grants; Dr. Vacca and Dr. Griffin by MRC program grant
(MC_PC_13030); Dr. Allison by the Evelyn Trust Grant. Dr.
Vacca was a fellow of Fondazione Umberto Veronesi (Italy).
The authors are members of the EPoS (Elucidating Pathways of Steatohepatitis) consortium funded by the Horizon 2020 Framework Program of the European Union
under Grant Agreement 634413.
ACC
ACL
ACSL
ACOX
ADP
Apo
ARA
AT
ALT
ATP
ChREBP
CPT-1
CVD
CYP4A
DNL
EPA
FA
FAS
FATP
FFA
FAO
FPLD3
HCC
HFD
HL
HODE
HSL
Insig1
IR
IRS2
IL
LCAD
LPL
MetS
MTP
NAFLD
NASH
OXPHOS
PCSK9
PEPCK
PG
PGC1
PPARs
PPREs
PUFAs
ROS
RXRs
SPARMs
SREBP1
TG
TZDs
UCPs
VLDL
acetyl-CoA carboxylase
ATP citrate lyase
acyl-CoA-synthetase
acyl-CoA oxidase
adenosine diphosphate
apolipoprotein
arachidonic acid
adipose tissue
alanine aminotransferase
adenosine triphosphate
carbohydrate response element-binding protein
carnitine palmityl transferase 1
cardiovascular disease
cytochrome P450 4A
de novo lipogenesis
eicosapentaenoic acid
fatty acid
fatty acid synthase
fatty acid binding protein
free fatty acid
fatty acid oxidation
familial partial lipodystrophy type 3
hepatocellular carcinoma
high-fat diet
hepatic lipase
hydroxyoctadecanoic acid
hormone-sensitive lipase
insulin-induced gene 1
insulin resistance
insulin receptor substrate 2
interleukin
long chain acyl-CoA dehydrogenase
lipoprotein lipase
metabolic syndrome
microsomal transfer protein
nonalcoholic fatty liver disease
nonalcoholic steatohepatitis
oxidative phosphorylation
proprotein convertase subtilisin/type 9
phosphoenolpyruvate carboxykinase
prostaglandin
peroxisome proliferator-activated receptor γ
coactivator 1
peroxisome proliferator-activated receptors
PPAR response elements
polyunsaturated fatty acids
reactive oxygen species
retinoid X receptors
selective PPARγ modulators
sterol regulatory element binding protein 1
triglyceride
thiazolidinediones
uncoupling proteins
very-low-density lipoprotein
Seminars in Liver Disease
Vol. 35
No. 3/2015
References
1 Matteoni CA, Younossi ZM, Gramlich T, Boparai N, Liu YC,
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
McCullough AJ. Nonalcoholic fatty liver disease: a spectrum of
clinical and pathological severity. Gastroenterology 1999;116(6):
1413–1419
Cohen JC, Horton JD, Hobbs HH. Human fatty liver disease: old
questions and new insights. Science 2011;332(6037):1519–1523
Adams LA, Angulo P, Lindor KD. Nonalcoholic fatty liver disease.
CMAJ 2005;172(7):899–905
Browning JD, Szczepaniak LS, Dobbins R, et al. Prevalence of
hepatic steatosis in an urban population in the United States:
impact of ethnicity. Hepatology 2004;40(6):1387–1395
Browning JD, Horton JD. Molecular mediators of hepatic steatosis
and liver injury. J Clin Invest 2004;114(2):147–152
Korenblat KM, Fabbrini E, Mohammed BS, Klein S. Liver, muscle,
and adipose tissue insulin action is directly related to intrahepatic triglyceride content in obese subjects. Gastroenterology
2008;134(5):1369–1375
Postic C, Girard J. The role of the lipogenic pathway in the
development of hepatic steatosis. Diabetes Metab 2008;34(6 Pt
2):643–648
He W, Barak Y, Hevener A, et al. Adipose-specific peroxisome
proliferator-activated receptor gamma knockout causes insulin
resistance in fat and liver but not in muscle. Proc Natl Acad Sci U S
A 2003;100(26):15712–15717
Medina-Gomez G, Gray SL, Yetukuri L, et al. PPAR gamma 2
prevents lipotoxicity by controlling adipose tissue expandability
and peripheral lipid metabolism. PLoS Genet 2007;3(4):e64
Rodriguez-Cuenca S, Carobbio S, Vidal-Puig A. Ablation of Pparg2
impairs lipolysis and reveals murine strain differences in lipolytic
responses. FASEB J 2012;26(5):1835–1844
Kempinska-Podhorodecka A, Krawczyk M, Klak M, et al. Healthy
PNPLA3 risk allele carriers present with unexpected body fat
composition. A study of one thousand subjects. J Gastrointestin
Liver Dis 2014;23(1):33–37
Cusi K. Role of obesity and lipotoxicity in the development of
nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology 2012;142(4):711–725.e6
Virtue S, Vidal-Puig A. Adipose tissue expandability, lipotoxicity
and the Metabolic Syndrome—an allostatic perspective. Biochim
Biophys Acta 2010;1801(3):338–349
Musso G, Cassader M, De Michieli F, Rosina F, Orlandi F, Gambino
R. Nonalcoholic steatohepatitis versus steatosis: adipose tissue
insulin resistance and dysfunctional response to fat ingestion
predict liver injury and altered glucose and lipoprotein metabolism. Hepatology 2012;56(3):933–942
Kraegen EW, Clark PW, Jenkins AB, Daley EA, Chisholm DJ, Storlien
LH. Development of muscle insulin resistance after liver insulin
resistance in high-fat-fed rats. Diabetes 1991;40(11):1397–1403
Rolo AP, Teodoro JS, Palmeira CM. Role of oxidative stress in the
pathogenesis of nonalcoholic steatohepatitis. Free Radic Biol Med
2012;52(1):59–69
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
Vacca et al.
17 Pessayre D, Fromenty B. NASH: a mitochondrial disease. J Hepatol
36 Sampath H, Miyazaki M, Dobrzyn A, Ntambi JM. Stearoyl-CoA
2005;42(6):928–940
Sanyal AJ, Campbell-Sargent C, Mirshahi F, et al. Nonalcoholic
steatohepatitis: association of insulin resistance and mitochondrial abnormalities. Gastroenterology 2001;120(5):1183–1192
Fan CY, Pan J, Usuda N, Yeldandi AV, Rao MS, Reddy JK. Steatohepatitis, spontaneous peroxisome proliferation and liver tumors
in mice lacking peroxisomal fatty acyl-CoA oxidase. Implications
for peroxisome proliferator-activated receptor alpha natural
ligand metabolism. J Biol Chem 1998;273(25):15639–15645
Sanyal AJ, Chalasani N, Kowdley KV, et al; NASH CRN. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N
Engl J Med 2010;362(18):1675–1685
Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD,
Parks EJ. Sources of fatty acids stored in liver and secreted via
lipoproteins in patients with nonalcoholic fatty liver disease.
J Clin Invest 2005;115(5):1343–1351
Adiels M, Taskinen MR, Packard C, et al. Overproduction of large
VLDL particles is driven by increased liver fat content in man.
Diabetologia 2006;49(4):755–765
Fabbrini E, Mohammed BS, Magkos F, Korenblat KM, Patterson
BW, Klein S. Alterations in adipose tissue and hepatic lipid
kinetics in obese men and women with nonalcoholic fatty liver
disease. Gastroenterology 2008;134(2):424–431
Horton JD, Shimano H, Hamilton RL, Brown MS, Goldstein JL.
Disruption of LDL receptor gene in transgenic SREBP-1a mice
unmasks hyperlipidemia resulting from production of lipid-rich
VLDL. J Clin Invest 1999;103(7):1067–1076
Duivenvoorden I, Teusink B, Rensen PC, Romijn JA, Havekes LM,
Voshol PJ. Apolipoprotein C3 deficiency results in diet-induced
obesity and aggravated insulin resistance in mice. Diabetes 2005;
54(3):664–671
Lee HY, Birkenfeld AL, Jornayvaz FR, et al. Apolipoprotein CIII
overexpressing mice are predisposed to diet-induced hepatic
steatosis and hepatic insulin resistance. Hepatology 2011;
54(5):1650–1660
Caron S, Verrijken A, Mertens I, et al. Transcriptional activation of
apolipoprotein CIII expression by glucose may contribute to
diabetic dyslipidemia. Arterioscler Thromb Vasc Biol 2011;
31(3):513–519
Chen M, Breslow JL, Li W, Leff T. Transcriptional regulation of the
apoC-III gene by insulin in diabetic mice: correlation with
changes in plasma triglyceride levels. J Lipid Res 1994;35(11):
1918–1924
Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by
nutrients and inflammation. J Clin Invest 2008;118(9):
2992–3002
Mittendorfer B, Magkos F, Fabbrini E, Mohammed BS, Klein S.
Relationship between body fat mass and free fatty acid kinetics in
men and women. Obesity (Silver Spring) 2009;17(10):
1872–1877
Anstee QM. Animal models in nonalcoholic steatohepatitis research: utility and clinical translation. Liver Int 2011;31(4):
440–442
Miyazaki M, Flowers MT, Sampath H, et al. Hepatic stearoyl-CoA
desaturase-1 deficiency protects mice from carbohydrate-induced adiposity and hepatic steatosis. Cell Metab 2007;6(6):
484–496
Bergheim I, Weber S, Vos M, et al. Antibiotics protect against
fructose-induced hepatic lipid accumulation in mice: role of
endotoxin. J Hepatol 2008;48(6):983–992
Feldstein AE, Canbay A, Guicciardi ME, Higuchi H, Bronk SF, Gores
GJ. Diet associated hepatic steatosis sensitizes to Fas mediated
liver injury in mice. J Hepatol 2003;39(6):978–983
Kennedy AR, Pissios P, Otu H, et al. A high-fat, ketogenic diet
induces a unique metabolic state in mice. Am J Physiol Endocrinol
Metab 2007;292(6):E1724–E1739
desaturase-1 mediates the pro-lipogenic effects of dietary saturated fat. J Biol Chem 2007;282(4):2483–2493
Romestaing C, Piquet MA, Bedu E, et al. Long term highly
saturated fat diet does not induce NASH in Wistar rats. Nutr
Metab (Lond) 2007;4:4
Newberry EP, Xie Y, Kennedy SM, Luo J, Davidson NO. Protection
against Western diet-induced obesity and hepatic steatosis in
liver fatty acid-binding protein knockout mice. Hepatology 2006;
44(5):1191–1205
Pfluger PT, Herranz D, Velasco-Miguel S, Serrano M, Tschöp MH.
Sirt1 protects against high-fat diet-induced metabolic damage.
Proc Natl Acad Sci U S A 2008;105(28):9793–9798
Singh R, Wang Y, Xiang Y, Tanaka KE, Gaarde WA, Czaja MJ.
Differential effects of JNK1 and JNK2 inhibition on murine
steatohepatitis and insulin resistance. Hepatology 2009;49(1):
87–96
Tetri LH, Basaranoglu M, Brunt EM, Yerian LM, NeuschwanderTetri BA. Severe NAFLD with hepatic necroinflammatory changes
in mice fed trans fats and a high-fructose corn syrup equivalent.
Am J Physiol Gastrointest Liver Physiol 2008;295(5):G987–G995
Koppe SW, Elias M, Moseley RH, Green RM. Trans fat feeding
results in higher serum alanine aminotransferase and increased
insulin resistance compared with a standard murine high-fat diet.
Am J Physiol Gastrointest Liver Physiol 2009;297(2):G378–G384
Obara N, Fukushima K, Ueno Y, et al. Possible involvement and the
mechanisms of excess trans-fatty acid consumption in severe
NAFLD in mice. J Hepatol 2010;53(2):326–334
Sekiya M, Yahagi N, Matsuzaka T, et al. Polyunsaturated fatty acids
ameliorate hepatic steatosis in obese mice by SREBP-1 suppression. Hepatology 2003;38(6):1529–1539
Baumgardner JN, Shankar K, Hennings L, Badger TM, Ronis MJ. A
new model for nonalcoholic steatohepatitis in the rat utilizing
total enteral nutrition to overfeed a high-polyunsaturated fat
diet. Am J Physiol Gastrointest Liver Physiol 2008;294(1):
G27–G38
Denechaud PD, Bossard P, Lobaccaro JM, et al. ChREBP, but not
LXRs, is required for the induction of glucose-regulated genes in
mouse liver. J Clin Invest 2008;118(3):956–964
Repa JJ, Liang G, Ou J, et al. Regulation of mouse sterol regulatory
element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRalpha and LXRbeta. Genes Dev 2000;14(22):2819–2830
Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the
complete program of cholesterol and fatty acid synthesis in the
liver. J Clin Invest 2002;109(9):1125–1131
Li Y, Xu S, Mihaylova MM, et al. AMPK phosphorylates and
inhibits SREBP activity to attenuate hepatic steatosis and atherosclerosis in diet-induced insulin-resistant mice. Cell Metab 2011;
13(4):376–388
Takaishi K, Duplomb L, Wang MY, Li J, Unger RH. Hepatic insig-1
or -2 overexpression reduces lipogenesis in obese Zucker diabetic
fatty rats and in fasted/refed normal rats. Proc Natl Acad Sci U S A
2004;101(18):7106–7111
Engelking LJ, Kuriyama H, Hammer RE, et al. Overexpression of
Insig-1 in the livers of transgenic mice inhibits SREBP processing
and reduces insulin-stimulated lipogenesis. J Clin Invest 2004;
113(8):1168–1175
Shimomura I, Matsuda M, Hammer RE, Bashmakov Y, Brown MS,
Goldstein JL. Decreased IRS-2 and increased SREBP-1c lead to
mixed insulin resistance and sensitivity in livers of lipodystrophic and ob/ob mice. Mol Cell 2000;6(1):77–86
Ide T, Shimano H, Yahagi N, et al. SREBPs suppress IRS-2mediated insulin signalling in the liver. Nat Cell Biol 2004;6(4):
351–357
Towle HC, Kaytor EN, Shih HM. Regulation of the expression of
lipogenic enzyme genes by carbohydrate. Annu Rev Nutr 1997;
17:405–433
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
34
35
37
38
39
40
41
42
43
44
45
46
47
48
49
50
51
52
53
54
Seminars in Liver Disease
Vol. 35
No. 3/2015
259
260
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
Vacca et al.
55 Iizuka K, Horikawa Y. ChREBP: a glucose-activated transcription
56
57
58
59
60
61
62
63
64
65
66
67
68
69
70
71
72
73
74
75
76
factor involved in the development of metabolic syndrome.
Endocr J 2008;55(4):617–624
Dentin R, Benhamed F, Hainault I, et al. Liver-specific inhibition of
ChREBP improves hepatic steatosis and insulin resistance in ob/
ob mice. Diabetes 2006;55(8):2159–2170
Benhamed F, Denechaud PD, Lemoine M, et al. The lipogenic
transcription factor ChREBP dissociates hepatic steatosis from
insulin resistance in mice and humans. J Clin Invest 2012;122(6):
2176–2194
Aarsland A, Wolfe RR. Hepatic secretion of VLDL fatty acids during
stimulated lipogenesis in men. J Lipid Res 1998;39(6):1280–1286
Korchak HM. Regulation of hepatic lipogenesis. Tufts Folia Med
1962;8:134–143
Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, Mangelsdorf DJ.
Anatomical profiling of nuclear receptor expression reveals a
hierarchical transcriptional network. Cell 2006;126(4):789–799
Mangelsdorf DJ, Thummel C, Beato M, et al. The nuclear receptor
superfamily: the second decade. Cell 1995;83(6):835–839
Mangelsdorf DJ, Evans RM. The RXR heterodimers and orphan
receptors. Cell 1995;83(6):841–850
Christodoulides C, Vidal-Puig A. PPARs and adipocyte function.
Mol Cell Endocrinol 2010;318(1-2):61–68
Atherton HJ, Gulston MK, Bailey NJ, et al. Metabolomics of the
interaction between PPAR-alpha and age in the PPAR-alpha-null
mouse. Mol Syst Biol 2009;5:259
Le May C, Pineau T, Bigot K, Kohl C, Girard J, Pégorier JP. Reduced
hepatic fatty acid oxidation in fasting PPARalpha null mice is due
to impaired mitochondrial hydroxymethylglutaryl-CoA synthase
gene expression. FEBS Lett 2000;475(3):163–166
Vu-Dac N, Gervois P, Jakel H, et al. Apolipoprotein A5, a crucial
determinant of plasma triglyceride levels, is highly responsive to
peroxisome proliferator-activated receptor alpha activators. J Biol
Chem 2003;278(20):17982–17985
Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, et al. PPARalpha
and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene.
EMBO J 1996;15(19):5336–5348
Wolfrum C, Borrmann CM, Borchers T, Spener F. Fatty acids and
hypolipidemic drugs regulate peroxisome proliferator-activated
receptors alpha - and gamma-mediated gene expression via liver
fatty acid binding protein: a signaling path to the nucleus. Proc
Natl Acad Sci U S A 2001;98(5):2323–2328
Martin G, Schoonjans K, Lefebvre AM, Staels B, Auwerx J. Coordinate regulation of the expression of the fatty acid transport
protein and acyl-CoA synthetase genes by PPARalpha and PPARgamma activators. J Biol Chem 1997;272(45):28210–28217
Fruchart JC. Peroxisome proliferator-activated receptor-alpha
(PPARalpha): at the crossroads of obesity, diabetes and cardiovascular disease. Atherosclerosis 2009;205(1):1–8
Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leitersdorf E,
Fruchart JC. Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation 1998;98(19):2088–2093
Vacca M, Degirolamo C, Mariani-Costantini R, Palasciano G,
Moschetta A. Lipid-sensing nuclear receptors in the pathophysiology and treatment of the metabolic syndrome. Wiley Interdiscip Rev Syst Biol Med 2011;3(5):562–587
Shah A, Rader DJ, Millar JS. The effect of PPAR-alpha agonism on
apolipoprotein metabolism in humans. Atherosclerosis 2010;
210(1):35–40
Ip E, Farrell GC, Robertson G, Hall P, Kirsch R, Leclercq I. Central
role of PPARalpha-dependent hepatic lipid turnover in dietary
steatohepatitis in mice. Hepatology 2003;38(1):123–132
Abdelmegeed MA, Yoo SH, Henderson LE, Gonzalez FJ, Woodcroft
KJ, Song BJ. PPARalpha expression protects male mice from high
fat-induced nonalcoholic fatty liver. J Nutr 2011;141(4):603–610
Su Q, Baker C, Christian P, et al. Hepatic mitochondrial and ER
stress induced by defective PPARα signaling in the pathogenesis
Seminars in Liver Disease
Vol. 35
No. 3/2015
77
78
79
80
81
82
83
84
85
86
87
88
89
90
91
92
93
of hepatic steatosis. Am J Physiol Endocrinol Metab 2014;
306(11):E1264–E1273
Chan SM, Sun RQ, Zeng XY, et al. Activation of PPARα ameliorates
hepatic insulin resistance and steatosis in high fructose-fed mice
despite increased endoplasmic reticulum stress. Diabetes 2013;
62(6):2095–2105
Stienstra R, Mandard S, Patsouris D, Maass C, Kersten S, Müller M.
Peroxisome proliferator-activated receptor alpha protects
against obesity-induced hepatic inflammation. Endocrinology
2007;148(6):2753–2763
Shiri-Sverdlov R, Wouters K, van Gorp PJ, et al. Early diet-induced
non-alcoholic steatohepatitis in APOE2 knock-in mice and its
prevention by fibrates. J Hepatol 2006;44(4):732–741
Laurin J, Lindor KD, Crippin JS, et al. Ursodeoxycholic acid or
clofibrate in the treatment of non-alcohol-induced steatohepatitis: a pilot study. Hepatology 1996;23(6):1464–1467
Fernández-Miranda C, Pérez-Carreras M, Colina F, López-Alonso
G, Vargas C, Solís-Herruzo JA. A pilot trial of fenofibrate for the
treatment of non-alcoholic fatty liver disease. Dig Liver Dis 2008;
40(3):200–205
Barish GD, Narkar VA, Evans RM. PPAR delta: a dagger in the heart
of the metabolic syndrome. J Clin Invest 2006;116(3):590–597
Grimaldi PA. Regulatory functions of PPARbeta in metabolism:
implications for the treatment of metabolic syndrome. Biochim
Biophys Acta 2007;1771(8):983–990
Berger J, Leibowitz MD, Doebber TW, et al. Novel peroxisome
proliferator-activated receptor (PPAR) gamma and PPARdelta
ligands produce distinct biological effects. J Biol Chem 1999;
274(10):6718–6725
Coleman JD, Prabhu KS, Thompson JT, et al. The oxidative stress
mediator 4-hydroxynonenal is an intracellular agonist of the
nuclear receptor peroxisome proliferator-activated receptorbeta/delta (PPARbeta/delta). Free Radic Biol Med 2007;42(8):
1155–1164
Wang YX, Lee CH, Tiep S, et al. Peroxisome-proliferator-activated
receptor delta activates fat metabolism to prevent obesity. Cell
2003;113(2):159–170
Roberts LD, Hassall DG, Winegar DA, Haselden JN, Nicholls AW,
Griffin JL. Increased hepatic oxidative metabolism distinguishes
the action of Peroxisome proliferator-activated receptor delta
from Peroxisome proliferator-activated receptor gamma in the
ob/ob mouse. Genome Med 2009;1(12):115
Barroso E, Rodríguez-Calvo R, Serrano-Marco L, et al. The PPARβ/δ
activator GW501516 prevents the down-regulation of AMPK
caused by a high-fat diet in liver and amplifies the PGC-1α-Lipin
1-PPARα pathway leading to increased fatty acid oxidation.
Endocrinology 2011;152(5):1848–1859
Liu S, Hatano B, Zhao M, et al. Role of peroxisome proliferatoractivated receptor delta/beta in hepatic metabolic regulation.
J Biol Chem 2011;286(2):1237–1247
Qin X, Xie X, Fan Y, et al. Peroxisome proliferator-activated
receptor-delta induces insulin-induced gene-1 and suppresses
hepatic lipogenesis in obese diabetic mice. Hepatology 2008;
48(2):432–441
Serrano-Marco L, Barroso E, El Kochairi I, et al. The peroxisome
proliferator-activated receptor (PPAR) β/δ agonist GW501516
inhibits IL-6-induced signal transducer and activator of transcription 3 (STAT3) activation and insulin resistance in human
liver cells. Diabetologia 2012;55(3):743–751
Nagasawa T, Inada Y, Nakano S, et al. Effects of bezafibrate, PPAR
pan-agonist, and GW501516, PPARdelta agonist, on development
of steatohepatitis in mice fed a methionine- and choline-deficient
diet. Eur J Pharmacol 2006;536(1-2):182–191
Vu-Dac N, Chopin-Delannoy S, Gervois P, et al. The nuclear
receptors peroxisome proliferator-activated receptor alpha and
Rev-erbalpha mediate the species-specific regulation of apolipoprotein A-I expression by fibrates. J Biol Chem 1998;273(40):
25713–25720
Fatty Acid and Glucose Sensors in Hepatic Lipid Metabolism
94 Tanaka T, Yamamoto J, Iwasaki S, et al. Activation of peroxisome
95
96
97
98
99
100
101
102
103
104
105
106
107
108
109
110
proliferator-activated receptor delta induces fatty acid betaoxidation in skeletal muscle and attenuates metabolic syndrome.
Proc Natl Acad Sci U S A 2003;100(26):15924–15929
Leibowitz MD, Fiévet C, Hennuyer N, et al. Activation of PPARdelta
alters lipid metabolism in db/db mice. FEBS Lett 2000;473(3):
333–336
Oliver WR Jr, Shenk JL, Snaith MR, et al. A selective peroxisome
proliferator-activated receptor delta agonist promotes reverse
cholesterol transport. Proc Natl Acad Sci U S A 2001;98(9):
5306–5311
Wallace JM, Schwarz M, Coward P, et al. Effects of peroxisome
proliferator-activated receptor alpha/delta agonists on HDL-cholesterol in vervet monkeys. J Lipid Res 2005;46(5):1009–1016
Kostadinova R, Montagner A, Gouranton E, et al. GW501516activated PPARβ/δ promotes liver fibrosis via p38-JNK MAPKinduced hepatic stellate cell proliferation. Cell Biosci 2012;2(1):
34
Vacca M, Degirolamo C, Massafra V, et al. Nuclear receptors in
regenerating liver and hepatocellular carcinoma. Mol Cell Endocrinol 2013;368(1-2):108–119
Vacca M, D’Amore S, Graziano G, et al. Clustering nuclear receptors in liver regeneration identifies candidate modulators of
hepatocyte proliferation and hepatocarcinoma. PLoS ONE 2014;
9(8):e104449
Savage DB, Tan GD, Acerini CL, et al. Human metabolic syndrome
resulting from dominant-negative mutations in the nuclear
receptor peroxisome proliferator-activated receptor-gamma. Diabetes 2003;52(4):910–917
Barroso I, Gurnell M, Crowley VE, et al. Dominant negative
mutations in human PPARgamma associated with severe insulin
resistance, diabetes mellitus and hypertension. Nature 1999;
402(6764):880–883
Semple RK, Chatterjee VK, O’Rahilly S. PPAR gamma and human
metabolic disease. J Clin Invest 2006;116(3):581–589
Jeninga EH, Gurnell M, Kalkhoven E. Functional implications of
genetic variation in human PPARgamma. Trends Endocrinol
Metab 2009;20(8):380–387
Gawrieh S, Marion MC, Komorowski R, et al. Genetic variation in
the peroxisome proliferator activated receptor-gamma gene is
associated with histologically advanced NAFLD. Dig Dis Sci 2012;
57(4):952–957
Krey G, Braissant O, L’Horset F, et al. Fatty acids, eicosanoids, and
hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor
ligand assay. Mol Endocrinol 1997;11(6):779–791
Dyson JK, Anstee QM, McPherson S. Non-alcoholic fatty liver
disease: a practical approach to treatment. Frontline Gastroenterol 2014;5(4):277–286
Chao L, Marcus-Samuels B, Mason MM, et al. Adipose tissue is
required for the antidiabetic, but not for the hypolipidemic, effect
of thiazolidinediones. J Clin Invest 2000;106(10):1221–1228
Maeda N, Takahashi M, Funahashi T, et al. PPARgamma ligands
increase expression and plasma concentrations of adiponectin,
an adipose-derived protein. Diabetes 2001;50(9):2094–2099
Collino M, Aragno M, Castiglia S, et al. Pioglitazone improves lipid
and insulin levels in overweight rats on a high cholesterol and
111
112
113
114
115
116
117
118
119
120
121
122
123
124
125
Vacca et al.
fructose diet by decreasing hepatic inflammation. Br J Pharmacol
2010;160(8):1892–1902
Hevener AL, Olefsky JM, Reichart D, et al. Macrophage PPAR
gamma is required for normal skeletal muscle and hepatic insulin
sensitivity and full antidiabetic effects of thiazolidinediones.
J Clin Invest 2007;117(6):1658–1669
Hevener AL, He W, Barak Y, et al. Muscle-specific Pparg deletion
causes insulin resistance. Nat Med 2003;9(12):1491–1497
Norris AW, Chen L, Fisher SJ, et al. Muscle-specific PPARgammadeficient mice develop increased adiposity and insulin resistance
but respond to thiazolidinediones. J Clin Invest 2003;112(4):
608–618
Lee YJ, Ko EH, Kim JE, et al. Nuclear receptor PPARγ-regulated
monoacylglycerol O-acyltransferase 1 (MGAT1) expression is
responsible for the lipid accumulation in diet-induced
hepatic steatosis. Proc Natl Acad Sci U S A 2012;109(34):
13656–13661
Morán-Salvador E, López-Parra M, García-Alonso V, et al. Role for
PPARγ in obesity-induced hepatic steatosis as determined by
hepatocyte- and macrophage-specific conditional knockouts.
FASEB J 2011;25(8):2538–2550
Yu S, Matsusue K, Kashireddy P, et al. Adipocyte-specific gene
expression and adipogenic steatosis in the mouse liver due to
peroxisome proliferator-activated receptor gamma1 (PPARgamma1) overexpression. J Biol Chem 2003;278(1):498–505
Cheng AY, Fantus IG. Oral antihyperglycemic therapy for type 2
diabetes mellitus. CMAJ 2005;172(2):213–226
Finck BN, Kelly DP. PGC-1 coactivators: inducible regulators of
energy metabolism in health and disease. J Clin Invest 2006;
116(3):615–622
Leone TC, Lehman JJ, Finck BN, et al. PGC-1alpha deficiency causes
multi-system energy metabolic derangements: muscle dysfunction, abnormal weight control and hepatic steatosis. PLoS Biol
2005;3(4):e101
Estall JL, Kahn M, Cooper MP, et al. Sensitivity of lipid metabolism
and insulin signaling to genetic alterations in hepatic peroxisome
proliferator-activated receptor-gamma coactivator-1alpha expression. Diabetes 2009;58(7):1499–1508
Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, Li X.
Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab
2009;9(4):327–338
Lin J, Yang R, Tarr PT, et al. Hyperlipidemic effects of dietary
saturated fats mediated through PGC-1beta coactivation of
SREBP. Cell 2005;120(2):261–273
Lelliott CJ, Medina-Gomez G, Petrovic N, et al. Ablation of PGC1beta results in defective mitochondrial activity, thermogenesis,
hepatic function, and cardiac performance. PLoS Biol 2006;4(11):
e369
Sonoda J, Mehl IR, Chong LW, Nofsinger RR, Evans RM. PGC-1beta
controls mitochondrial metabolism to modulate circadian activity, adaptive thermogenesis, and hepatic steatosis. Proc Natl Acad
Sci U S A 2007;104(12):5223–5228
Bellafante E, Murzilli S, Salvatore L, Latorre D, Villani G, Moschetta
A. Hepatic-specific activation of peroxisome proliferator-activated receptor γ coactivator-1β protects against steatohepatitis.
Hepatology 2013;57(4):1343–1356
Seminars in Liver Disease
Vol. 35
No. 3/2015
261