You are on page 1of 8

Int J Clin Exp Med 2014;7(7):1624-1631

www.ijcem.com /ISSN:1940-5901/IJCEM0000962

Review Article
Signal transductions and nonalcoholic fatty liver: a
mini-review
Lu Zeng, Wai J Tang, Jin J Yin, Bei J Zhou

Department of Pharmacy, Zhu-Jiang Hospital, Southern Medical University, Guangzhou 510280, China
Received November 21, 2013; Accepted May 31, 2014; Epub July 15, 2014; Published July 30, 2014

Abstract: Nonalcoholic fatty liver disease (NAFLD) is a common liver disease, and the incidence increases year by
year. The pathogenesis of NAFLD is correlated with insulin resistant (IR), and oxidative stress which induces varied
inflammatory cytokines (TNF-, IL-1, IL-6, etc). Different signal transductions such as MAPK, NF-B, AMPK, JAK2/
STAT3, PPAR, PI3K/Akt, TLR were activated by the pathogenic factors to regulate correlative reactions. Thus, in-
depth study of the signal transductions will probably provide new suitable solutions for the prevention and therapy
of NAFLD.

Keywords: Nonalcoholic fatty liver disease, pathogenesis, signal transduction

Nonalcoholic Fatty Liver Disease which was year, and their ages are to be younger, there-
coined by Ludwing in 1980 early is a clinical fore, NAFLD has become a global public health
syndrome characterized by the excessive fat problem. So far the pathogenesis of NAFLD has
accumulation in the liver and hepatic cellular not been fully clarified, and currently, a variety
degeneration even in the absence of alcohol of factors based on the insulin-oxidative stress
consumption [1]. Defined as a genetic-environ- injury are considered to be involved in it, but its
mental-metabolic stress-related disease, NAF- intracellular signal transduction mechanisms
LD encompasses a spectrum of disease from are not clear yet. As is known that the intracel-
simple steatosis to nonalcoholic steatohepati- lular signaling transduction is an important way
tis (NASH), which can progress to cirrhosis and for a numerous factors to stimulate cell chang-
hepatocellular carcinoma. NAFLD affects 10% es, in other words, the relevant causative fac-
to 24% of the general population in various tors play a role by the signal transmission within
countries and the prevalence has even been up the liver cells. Currently many factors such as
free fatty acids (FFAs), reactive oxidative stress
to 75 percent in obese people [2]. In United
(ROS), tumor necrosis factor- (TNF-), interleu-
States it translates to approximately 30.1 mil-
kin (IL)-6 that cause insulin resistance (IR),
lion obese people affected with steatosis and
hepatocyte fat accumulation and cellular injury
8.6 million with steatohepatitis [3]. Hyperlipide-
are involved in the pathologic processes of
mia (hypertriglyceridemia and/or hypercholes- NAFLD by directly activating the c-Jun N-terminal
terolemia), which is frequently associated with kinase (JNK) [5]. Otherwise, signaling pathways
both obesity and type 2 diabetes, has been related with insulin resistance, oxidative stress
reported in 20% to 80% of patients with NASH and inflammation fibrosis include: nuclear fac-
[4]. There is increasing evidence that NAFLD tor B (NF-B), AMP-activated protein kinase
represents the hepatic component of a meta- (AMPK), Janus kinase/signal transducers and
bolic syndrome characterized by obesity, hyper- activators of transcription (JAK/STAT), peroxi-
insulinemia, peripheral insulin resistance, dia- some proliferator-activated-receptors (PPARs,
betes, hypertriglyceridemia, and hypertension. phosphatidylinositol 3-kinase/protein kinase B
In recent years, with the improvement of living (PI3K/Akt), Toll-like receptor (TLR). Blocking any
standard and lifestyle, especially the change of one of the above pathways would not be effec-
the diet structure and decrease in physical tive in the prevention and treatment of NAFLD
activity, the NAFLD patients increase year by [6-11].
Nonalcoholic fatty liver disease in signal transductions

Figure 1. The pathogenesis of NAFLD: two-hit model.

Pathogenesis of NAFLD more hit by fibrosis factors (such as TGF-)


results in the synthesis of hepatocyte extracel-
The pathogenesis of NAFLD is not a simple lular matrix is greater than the degradation,
mechanism, the most widespread and prevail- thus forming a progressive fibrosis [18].
ing theory is the so-called two-hit model Although a variety of factors are involved in the
(Figure 1). The first hit is insulin resistant(IR) pathogenesis of NAFLD, the development of
leading to hepatic fat accumulation; on this fatty liver is a highly integrated process. Now
basis a large number of adipokines (leptin, adi- its clear that intracellular signaling transduc-
ponectin, resistin) regulate free fatty acids tion is an important way for a variety of factors
(FFAs) to induce ROS injury is to be the second to stimulate cell changes. Namely, the relevant
hit [3, 12-15]; ROS could activate Fas ligand/ causative factors play an important role by the
Fas system, and progress to lead to structural signal transmission within the liver cells, so it
protein of the Fas death zone to raise the down- will be helpful for the prevention and cure of
stream caspase family members to form the NAFLD by in-depth study of the signal transduc-
protease procascade reaction and then results tions in liver cells about the pathogenesis of
in cellular disorganization and apoptosis; NAFLD.
Whats more, the apoptotic hepatocytes could
form the aggregation of inflammatory cells, Signal transductions and NAFLD
thereby inducing a variety of inflammatory cyto-
kines (TNF-, IL-1, IL-6, IL-8, IL-18, monocyte MAPK and NAFLD
chemoattractant protein (MCP-1), etc.) directly
mediate fibrosis to induce inflammation of the Mitogen-activated protein kinase (MAPK) is a
liver and steatohepatitis, the process above is class of serine/threonine protein kinase widely
inflammatory-necrotic circulation [16, 17]; One existed in mammalian cells and mediates sig-

1625 Int J Clin Exp Med 2014;7(7):1624-1631


Nonalcoholic fatty liver disease in signal transductions

nal conduction from the cell surface to the JNK and p38 signaling pathways are involved in
endonuclear, which includes cell extracellular the pathological process of NAFLD.
signal-regulated kinases (ERKs), c-Jun amino-
terminal kinase (JNK) and p38MAPK. The ERKs NF-B and NAFLD
play a major role in the cellular response
induced by growth factor stimulation, and JNK Nuclear factor-B (NF-B) is a nuclear transcrip-
and p38 are related with stress and inflamma- tion factor widely present in varied cells, which
tion [19]. MAPK activation is involved in regula- could regulate a variety of cytokines involved in
tion of cell proliferation, differentiation, trans- inflammation, adhesion molecules and prote-
formation and apoptosis through phosphory- ase gene transcription in vivo, and it is closely
lation of nuclear transcription factors, cytoskel- related with inflammation. NF-B activation can
etal proteins and enzymes, which is closely promote the expression of inflammatory cyto-
related with mechanisms of inflammation, can- kines, while inflammatory factors, in turn, can
cer and many other diseases [20]. In recent further enhance the activity of NF-B, to make
years, many researches have indicated that the inflammation worse [25]. Cytokines (TNF-,
TNF-, IL-1, etc.), growth factors (such as insu-
JNK was closely related with IR; And inflamma-
lin), immune receptors, the medium, stress
tory cytokines such as tumor necrosis factor
response (oxygenation, etc.), bacteria and prod-
(TNF-), free fatty acids (FFAs), oxidative stress
ucts (lipopolysaccharide, etc.), viruses and the
that could lead to IR, hepatocyte fat accumula-
product, biological xenobiotics, environmental
tion and cell injury were involved in the patho-
hazards, and other factors could induce NF-B
genesis of NAFLD rat model by activating the
activation [25], initiating transcription of many
JNK [5, 21]. Dynamic observation of the JNK
genes such as TNF-, IL-1, IL-6, IL-8 after activa-
signaling pathway protein expression in rat liver
tion. These gene products regulated by NF-B
tissue and the generated impact and mecha-
are involved in inflammation of the liver, liver
nism of IR in the process of the formation of
fibrosis, liver regeneration and apoptosis. In
high-fat-diet induced NAFLD from the first
addition, FFAs, ROS initiate the activation of the
week, drawing that the high-fat-diet rat liver tis-
transcription factor NF-B, which leads to
sue JNK1 protein expression levels were higher
increased pro-inflammatory cytokines (TNF-,
compared with the control group over the same
TGF-, IL-1, IL-6, IL-8) production, and insulin
period, and JNK1 protein expression and IR
resistance in the liver [4]. Several pro-inflam-
level was positively correlated from the end of
matory cytokines such as tumor necrosis
the second week, which indicated that high-fat-
factor- (TNF-), interleukin (IL)-1, IL-6, various
diet activated JNK1, diminished insulin signal-
adipocytokines, and several transcription fac-
ing, thus causing IR [22]. p38 signaling path-
tors and kinases such as c-Jun N-terminal
ways are involved in the cell inflammatory
kinase (JNK) and a kinase located proximal of
response and apoptosis process under stress
nuclear factor-B (NF-B) participate in the
conditions, which are related with the release
occurrence and development of the IR [6]. The
of a variety of inflammatory cytokines (such as
IR and inflammatory cytokines are related with
IL-1, TNF- and IL-6, etc.) after activation [23].
the pathogenesis of NAFLD. There were also
To study the action mechanism of an antioxi-
experiments confirmed that NAFLD rat liver tis-
dant in the high-fat-diet rats, Sinha-Hikim et al sue NF-B expression was significantly enhan-
[24] analyzed the phosphorylation of JNK and ced than the normal group [26]. Therefore, the
p38 signal level in the high-fat-diet-induced NF-B signaling pathway are probably involved
NAFLD rats, which confirmed a significant in the pathological process of NAFLD.
(P<0.05) increase in both phospho-JNK and
phospho-p38MAPK levels than the normal con- AMPK and NAFLD
trol group and significantly reduced after anti-
oxidants treatment, and drew that oxidative AMP-activated protein kinase (AMPK) is a het-
stress can promote activation of both p38MAPK erologous trimeric protein kinase activated by
and JNK, which through mitochondria-depen- adenosine monophosphate (AMP) and widely
dent intrinsic pathway signaling promotes exists in eukaryotic cells, which is a central reg-
apoptosis in various cell types. The pathogen- ulator of cellular energy balance and plays an
esis of NAFLD is related with IR and a variety of important role in fatty acid metabolism through
inflammatory cytokines, so it is known that the the fatty acid biosynthetic pathway. AMPK is

1626 Int J Clin Exp Med 2014;7(7):1624-1631


Nonalcoholic fatty liver disease in signal transductions

composed of , and subunit, is the cata- and liver) over the body by circulation and com-
lytic subunit controlled by AMP/ATP ratio, which bines with its receptors, which then interact
could be activated by stress stimuli caused by with Janus family protein tyrosine kinase/signal
any reduction in intracellular ATP that include transduction and activates transcription fac-
metabolic product, the oxidation stress, hypox- tors (JAK/STAT), mainly JAK2/STAT3 [33], to
ia and low sugar [27]; when the AMP/ATP ratio cause the related biological effects, and to
increases, AMPK phosphorylation activates a exhibit the function of diet control, energy
large number of downstream target molecule metabolism regulation and interfere with the
acetyl coenzyme A carboxylase (ACC), which role of insulin in the liver, thus decreasing tri-
reduce the use of ATP (inhibition of glycogen, glyceride (TG), elevating the insulin sensitivity
fat and cholesterol synthesis) and increase ATP of liver and peripheral tissues and reducing fat
production (promotion of fatty acid oxidation deposition [8]; When the receptor expression is
and glucose transporter) [28], so that the cell in dysfunction, JAK2/STAT3 signal transduction
catabolism increases. Sterol regulatory ele- disorder occurs, thus leptin could not play the
ment-binding proteins (SREBPs) are a family of biological effects to cause the leptin resis-
transcription factors that control the expres- tance. The NAFLD patients commonly have
sion of genes required for the biosynthesis of leptin resistance [34], thereby promoting lipid
cholesterol, fatty acids, triglycerides, and phos- synthesis in the liver and leading to the devel-
pholipids, and SREBP-1c preferentially controls opment of fatty liver. Bartek [35] et al. found
the expression of genes involved in triglyceride that leptin and free leptin receptor in the serum
synthesis and accumulation, such as fatty acid of patients with fatty liver were significantly
synthase (FAS) and ACC [29]. Therefore, AMPK increased, suggesting that leptin bound to its
activation suppresses the expression of ACC receptor may exist obstacles to cause leptin
and FAS via down-regulation of SREBP-1c [30]. resistance. The experimental results showed
AMPK activators, metformin, have been shown that leptin receptor mRNA and phosphorylation
to inhibit the expression of the SREPB-1c gene of JAK2/STAT3 level in NAFLD rat liver were
and to prevent the development of hepatic ste- lower than that in the control group [8]. To sum
atosis [31]. Adiponectin is a recently discovered up, leptin JAK2/STAT3 signal pathway is proba-
hormone against diabetes insulin resistance, bly involved in the pathological process of
which comes from the fat cells, has function of NAFLD.
regulating energy balance, glucose and fat
metabolism, and it is closely related with insu- PPARs and NAFLD
lin resistance and atherosclerosis hardening.
Peroxisome proliferator-activated receptors
Adiponectin is inhibited by TNF-, IL-6, resistin,
(PPARs) are ligand-activated transcription fac-
insulin, and stimulates mitochondrial -oxida-
tors of nuclear receptor family that exist three
tion by activating AMPK and inhibits lipogene-
subtypes, namely , , and . PPAR mainly dis-
sis by down-regulating SREBP-1c [28]. Yamauchi
tributes in the tissue with a high efficiency of
[32] et al. also believed that its signal transduc-
mitochondrial fatty acid oxidation, which highly
tion mechanisms were activating AMPK func-
expresses in the liver, and PPAR highly pres-
tion. In conclusion, AMPK is closely related with
ents in adipose tissue and the immune system.
insulin resistance and liver lipid content. A
PPARs regulate not only the expression of
number of experimental studies also confirmed
genes involved in fatty acid synthesis, oxida-
that phosphorylated AMPK protein expression
tion, and storage, but also participate in the
level in high-fat-diet-induced NAFLD was signifi-
molecular mechanism of altered metabolic
cantly lower than the normal control group [7,
homeostasis, such as is found in type 2 diabe-
24], which suggested that AMPK was involved
tes or obesity [9, 36]. PPAR agonist and PPAR
in the pathological process of NAFLD.
agonist are involved in diabetes mellitus and
JAK2/STAT3 and NAFLD dyslipidemia, and they play an important role in
insulin resistance [37]. Seo [38] et al found
Leptin is the main regulator of fat in the organ- PPAR agonists, especially a PPAR agonist,
ism. It is released from the fat tissue into blood, improved the histological and biochemical
then dispensed into the tissues (musculi skel- parameters in the NAFLD rat model by inducing
eti, adipose tissue, peripheral lymphoid tissue, fatty-acid metabolic enzymes. PPAR is consid-
central nervous system, gastrointestinal tract, ered to be the main regulator of fatty acid oxi-

1627 Int J Clin Exp Med 2014;7(7):1624-1631


Nonalcoholic fatty liver disease in signal transductions

dation: PPAR is activated in combination with TLR and NAFLD


polyunsaturated fatty acids, thereby it can
improve insulin resistance, reduce blood lipids, TLR (Toll-like receptor) family members are
promote -oxidation of fatty acids, inhibit lipo- characterized by highly evolutionarily con-
genesis gene expression, inhibit genes tran- served TIR domain (Toll/IL-1R domain) in the
scription related with inflammatory response, intracellular region, which could recognize var-
which are conducive to control NAFLD [39]. In ies of pathogen-associated molecular patterns
addition, PPAR is activated by adiponectin (PAMP) by the leucine repeat(leucine-rich
and could inhibit NF-kB pathway [28]. PPAR repeat LRR) Ribbon in the extracellular region.
enhances insulin action, FFA oxidation, adipo- Various TLR could induce a series of gene acti-
nectin secretion, and inhibits secretion of pro- vation through the same or a different signal
inflammatory cytokines [28], so PPAR could transduction pathway in pathogen invasion
improve NAFLD. However hepatic PPAR 2 minutes, resulting in cell secretion of pro-
expression is increased in high-fat-diet fed inflammatory cytokines and chemokines,
mice due to elevated rates of lipogenesis via upregulation of co-stimulatory factor, increase
the upregulation of de novo lipogenic genes of antigen-presenting ability, which cause sys-
FAS and ACC [40]. So it is still not conclusive as temic inflammatory response and the occur-
to whether the PPAR is beneficial or detrimen- rence of innate immune response [46].
tal. Thus, we could know PPARs play an impor- Knockout rats results showed that TLR4, TLR9
tant role in the pathogenesis of fatty liver. promoted the development of NAFLD [47].
TLR4 is LPS receptor, and LPS elevation in most
PI3K/Akt and NAFLD animal models of NAFLD causes liver steatosis,
hepatic insulin resistance, increased liver
Phosphatidylinositol 3-kinase (PI3K) pathway is
weight, so the TLR4-LPS is the key pathway to
one of the main signal transduction pathways
promote NAFLD development [48]. In addition,
of insulin action, and PKB/Akt (protein kinase
FFAs could also activate TLR4 [47]. TLR9 signal
B) as PI3K downstream kinase is the important
is related with the development of NASH, TLR9
serine/threonine kinase in this pathway which
missing rats present less steatohepatitis, insu-
is primarily responsible for the conduction of
lin resistance, and fiber reaction [49]. MyD88 is
the initial biological information by PI3K. PI3K/
an adapter protein of all TLRs except TLR3,
Akt signaling pathway as insulin downstream
which is related with expression of a variety of
molecular pathway, plays an important role in a
inflammatory cytokines and inflammatory che-
variety of biological processes such as cell
mokines, and TLR-MyD88 signaling pathway
metabolism, cell cycle regulation, cell growth,
could also activate JNK and NF-B signaling
apoptosis, glucose transporter, and it is closely
[47], while JNK and NF-B signals are involved
related with the development of IR [10]. In addi-
in the pathogenesis of NAFLD. TLR signal medi-
tion, the p85 subunit of PI3K could be com-
bined with the insulin receptor substrate, so ates the occurrence of steatosis, inflammation
close to the insulin receptor and is anchored in and fibrosis, which is closely related with the
the cell membrane, thereby activating the p110 incidence of NAFLD.
subunit to regulate fat cells and liver cells in the Conclusion
uptake of glucose through a series of signal
transduction [41]. Yuan J [42] et al. found that NAFLD is increasingly common in the crowd,
long-term insulin-stimulated HepG2 cells could the two-hit theory based on IR and oxidative
lower insulin signal transduction through the stress is the main theoretical basis to explain
PI3K signaling pathway to inhibit insulin signal- its pathogenesis. Based on the pathogenesis
ing, leading to insulin resistance, meanwhile it of NAFLD, it could be found that MAPK, NF-B,
has been confirmed that insulin signal could AMPK, JAK2/STAT3, PPARs, PI3K/Akt, TLR sig-
not pass in the direction of glucose uptake naling pathways are closely related with the
through the PI3K pathway and to cause IR if the incidence of NAFLD. In the NAFLD pathogene-
PI3K expression and activity reduce. Akt could sis, the same kind of stimulation could activate
suppress fatty acid oxidation gene expression, different signal pathways, whereas different
thereby regulating the process of hepatic glu- stimuli could activate a signal pathway (Figure
cose and lipid metabolism [43]. There were 2): Inflammatory cytokines (TNF-, TGF-, IL-1,
some study results showed that NAFLD rat liver IL-6, etc.) could activate JNK, p38 and NF-B
PI3K and Akt protein were significantly lower signaling pathways, in turn, the activation of
than the normal groups [44, 45]. these signaling pathways could increase the

1628 Int J Clin Exp Med 2014;7(7):1624-1631


Nonalcoholic fatty liver disease in signal transductions

Figure 2. The signal transductions in NAFLD.

expression of inflammatory cytokines; adipo- risk factors for progression of non-alcoholic


nectin could reduce fatty acid synthesis and fatty liver disease and steatohepatitis. Eur Rev
enhance fatty acid -oxidation via activation of Med Pharmacol Sci 2005; 9: 273-277.
AMP kinase, and then resist steatosis and [3] Yan E, Durazo F, Tong M, Hong K. Nonalcoholic
increase insulin sensitivity [50], but also could fatty liver disease: pathogenesis, identifica-
activate PPAR and inhibit NF-kB pathway [13]; tion, progression, and management. Nutr Rev
2007; 65: 376-384.
resistin could inhibit AMPK activity in liver and
[4] Duvnjak M, Lerotic I, Barsic N, Tomasic V, Vi-
skeletal muscle, impede insulin signaling PI3K/
rovic JL, Velagic V. Pathogenesis and manage-
Akt pathway and induce NF-B nuclear tran- ment issues for non-alcoholic fatty liver dis-
scription [28, 51, 52]; leptin plays biological ease. World J Gastroenterol 2007; 13: 4539-
effects through JAK2/STAT3, may also stimu- 4550.
late AMPK to increase in -oxidation [28]. [5] Kodama Y, Brenner DA. c-Jun N-terminal ki-
These signaling pathways are interconnected nase signaling in the pathogenesis of nonalco-
to form a network of signaling pathways, so holic fatty liver disease: Multiple roles in multi-
blocking any one of pathway could not be effec- ple steps. Hepatology 2009; 49: 6-8.
tive to prevent and treat the NAFLD. Therefore, [6] Tilg H, Moschen AR. Inflammatory mecha-
in-depth study of liver cells signaling in the nisms in the regulation of insulin resistance.
NAFLD pathogenesis process will contribute to Mol Med 2008; 14: 222-231.
the prevention and treatment of NAFLD. [7] Park KG, Min AK, Koh EH, Kim HS, Kim MO,
Park HS, Kim YD, Yoon TS, Jang BK, Hwang JS,
Address correspondence to: Dr. Bei J Zhou, Depart- Kim JB, Choi HS, Park JY, Lee IK, Lee KU. Al-
ment of Pharmacy, Zhu-Jiang Hospital, Southern pha-lipoic acid decreases hepatic lipogenesis
through adenosine monophosphate-activated
Medical University, Industrial Road, No. 253, Haizhu
protein kinase (AMPK)-dependent and AMPK-
District, Guangzhou 510280, China. Tel: 020-6164-
independent pathways. Hepatology 2008; 48:
3549; Fax: 020-84300639; E-mail: zhoubj163@
1477-1486.
163.com; zly13148@gmail.com
[8] Zheng P, Ji G, Ma Z, Liu T, Xin L, Wu H, Liang X,
Liu J. Therapeutic effect of puerarin on non-al-
References
coholic rat fatty liver by improving leptin signal
[1] Yeh MM, Brunt EM. Pathology of nonalcoholic transduction through JAK2/STAT3 pathways.
fatty liver disease. Am J Clin Pathol 2007; 128: Am J Chin Med 2009; 37: 69-83.
837-847. [9] Aleman G, Torres N, Tovar AR. [Peroxisome
[2] Miele L, Forgione A, Hernandez AP, Gabrieli proliferator-activated receptors (PPARs) in obe-
ML, Vero V, Di Rocco P, Greco AV, Gasbarrini G, sity and insulin resistance development]. Rev
Gasbarrini A, Grieco A. The natural history and Invest Clin 2004; 56: 351-367.

1629 Int J Clin Exp Med 2014;7(7):1624-1631


Nonalcoholic fatty liver disease in signal transductions

[10] Cantley LC. The phosphoinositide 3-kinase [25] Kaidashev IP. [NF-kB activation as a molecular
pathway. Science 2002; 296: 1655-1657. basis of pathological process by metabolic syn-
[11] Mencin A, Kluwe J, Schwabe RF. Toll-like recep- drome]. Fiziol Zh 2012; 58: 93-101.
tors as targets in chronic liver diseases. Gut [26] Leclercq IA, Farrell GC, Sempoux C, Dela PA,
2009; 58: 704-720. Horsmans Y. Curcumin inhibits NF-kappaB ac-
[12] Tiniakos DG, Vos MB, Brunt EM. Nonalcoholic tivation and reduces the severity of experimen-
fatty liver disease: pathology and pathogene- tal steatohepatitis in mice. J Hepatol 2004; 41:
sis. Annu Rev Pathol 2010; 5: 145-171. 926-934.
[13] Polyzos SA, Kountouras J, Zavos C, Tsiaousi E. [27] Zhang BB, Zhou G, Li C. AMPK: an emerging
The role of adiponectin in the pathogenesis drug target for diabetes and the metabolic syn-
and treatment of non-alcoholic fatty liver dis- drome. Cell Metab 2009; 9: 407-416.
ease. Diabetes Obes Metab 2010; 12: 365- [28] Musso G, Gambino R, Cassader M. Non-alco-
383. holic fatty liver disease from pathogenesis to
[14] Gentile CL, Frye MA, Pagliassotti MJ. Fatty ac- management: an update. Obes Rev 2010; 11:
ids and the endoplasmic reticulum in nonalco- 430-445.
holic fatty liver disease. Biofactors 2011; 37: [29] Yuan H, Shyy JY, Martins-Green M. Second-
8-16. hand smoke stimulates lipid accumulation in
the liver by modulating AMPK and SREBP-1. J
[15] Tilg H. Adipocytokines in nonalcoholic fatty liv-
Hepatol 2009; 51: 535-547.
er disease: key players regulating steatosis,
[30] Kohjima M, Higuchi N, Kato M, Kotoh K, Yoshi-
inflammation and fibrosis. Curr Pharm Des
moto T, Fujino T, Yada M, Yada R, Harada N,
2010; 16: 1893-1895.
Enjoji M, Takayanagi R, Nakamuta M. SREBP-
[16] Kneeman JM, Misdraji J, Corey KE. Secondary
1c, regulated by the insulin and AMPK signal-
causes of nonalcoholic fatty liver disease.
ing pathways, plays a role in nonalcoholic fatty
Therap Adv Gastroenterol 2012; 5: 199-207.
liver disease. Int J Mol Med 2008; 21: 507-
[17] Harmon RC, Tiniakos DG, Argo CK. Inflamma- 511.
tion in nonalcoholic steatohepatitis. Expert [31] Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-
Rev Gastroenterol Hepatol 2011; 5: 189-200. Melody J, Wu M, Ventre J, Doebber T, Fujii N,
[18] De Minicis S, Svegliati-Baroni G. Fibrogenesis Musi N, Hirshman MF, Goodyear LJ, Moller DE.
in nonalcoholic steatohepatitis. Expert Rev Role of AMP-activated protein kinase in mech-
Gastroenterol Hepatol 2011; 5: 179-187. anism of metformin action. J Clin Invest 2001;
[19] Chakrabarty S, Kondratick L. Insulin-like 108: 1167-1174.
growth factor binding protein-2 stimulates pro- [32] Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki
liferation and activates multiple cascades of H, Uchida S, Yamashita S, Noda M, Kita S, Ueki
the mitogen-activated protein kinase pathways K, Eto K, Akanuma Y, Froguel P, Foufelle F,
in NIH-OVCAR3 human epithelial ovarian can- Ferre P, Carling D, Kimura S, Nagai R, Kahn BB,
cer cells. Cancer Biol Ther 2006; 5: 189-197. Kadowaki T. Adiponectin stimulates glucose
[20] Cargnello M, Roux PP. Activation and function utilization and fatty-acid oxidation by activating
of the MAPKs and their substrates, the MAPK- AMP-activated protein kinase. Nat Med 2002;
activated protein kinases. Microbiol Mol Biol 8: 1288-1295.
Rev 2011; 75: 50-83. [33] Saxena NK, Ikeda K, Rockey DC, Friedman SL,
[21] Tarantino G, Caputi A. JNKs, insulin resistance Anania FA. Leptin in hepatic fibrosis: evidence
and inflammation: A possible link between for increased collagen production in stellate
NAFLD and coronary artery disease. World J cells and lean littermates of ob/ob mice. Hepa-
Gastroenterol 2011; 17: 3785-3794. tology 2002; 35: 762-771.
[22] Malhi H, Bronk SF, Werneburg NW, Gores GJ. [34] Chitturi S, Abeygunasekera S, Farrell GC,
Free fatty acids induce JNK-dependent hepato- Holmes-Walker J, Hui JM, Fung C, Karim R, Lin
cyte lipoapoptosis. J Biol Chem 2006; 281: R, Samarasinghe D, Liddle C, Weltman M,
12093-12101. George J. NASH and insulin resistance: Insulin
[23] Pearson G, Robinson F, Beers GT, Xu BE, hypersecretion and specific association with
Karandikar M, Berman K, Cobb MH. Mitogen- the insulin resistance syndrome. Hepatology
activated protein (MAP) kinase pathways: regu- 2002; 35: 373-379.
lation and physiological functions. Endocr Rev [35] Bartek J, Bartos J, Galuska J, Galuskova D, Ste-
2001; 22: 153-183. jskal D, Ivo K, Ehrmann J, Ehrman JJ, Chlup R.
[24] Sinha-Hikim I, Sinha-Hikim AP, Shen R, Kim HJ, Expression of ob gene coding the production of
French SW, Vaziri ND, Crum AC, Rajavashisth the hormone leptin in hepatocytes of liver with
TB, Norris KC. A novel cystine based antioxi- steatosis. Biomed Pap Med Fac Univ Palacky
dant attenuates oxidative stress and hepatic Olomouc Czech Repub 2001; 145: 15-20.
steatosis in diet-induced obese mice. Exp Mol [36] Ferre P. The biology of peroxisome proliferator-
Pathol 2011; 91: 419-428. activated receptors: relationship with lipid me-

1630 Int J Clin Exp Med 2014;7(7):1624-1631


Nonalcoholic fatty liver disease in signal transductions

tabolism and insulin sensitivity. Diabetes [45] Matsuda S, Kobayashi M, Kitagishi Y. Roles for
2004; 53 Suppl 1: S43-S50. PI3K/AKT/PTEN Pathway in Cell Signaling of
[37] Choi KC, Ryu OH, Lee KW, Kim HY, Seo JA, Kim Nonalcoholic Fatty Liver Disease. ISRN Endo-
SG, Kim NH, Choi DS, Baik SH, Choi KM. Effect crinol 2013; 2013: 472432.
of PPAR- and - agonist on the expression of [46] Seki E, Brenner DA. Toll-like receptors and
visfatin, adiponectin, and TNF- in visceral fat adaptor molecules in liver disease: update.
of OLETF rats. Biochem Bioph Res Co 2005; Hepatology 2008; 48: 322-335.
336: 747-753. [47] Miura K, Seki E, Ohnishi H, Brenner DA. Role of
[38] Seo YS, Kim JH, Jo NY, Choi KM, Baik SH, Park toll-like receptors and their downstream mole-
JJ, Kim JS, Byun KS, Bak YT, Lee CH, Kim A, cules in the development of nonalcoholic Fatty
Yeon JE. PPAR agonists treatment is effective liver disease. Gastroenterol Res Pract 2010;
in a nonalcoholic fatty liver disease animal 2010: 362847.
model by modulating fatty-acid metabolic en- [48] Soares JB, Pimentel-Nunes P, Roncon-Albu-
zymes. J Gastroenterol Hepatol 2008; 23: 102- querque R, Leite-Moreira A. The role of lipo-
109. polysaccharide/toll-like receptor 4 signaling in
[39] Videla LA, Pettinelli P. Misregulation of PPAR chronic liver diseases. Hepatol Int 2010; 4:
Functioning and Its Pathogenic Consequences 659-672.
Associated with Nonalcoholic Fatty Liver Dis- [49] Miura K, Kodama Y, Inokuchi S, Schnabl B,
ease in Human Obesity. PPAR Res 2012; 2012: Aoyama T, Ohnishi H, Olefsky JM, Brenner DA,
107434. Seki E. Toll-like receptor 9 promotes steato-
[40] Ables GP. Update on ppargamma and nonalco- hepatitis by induction of interleukin-1beta in
holic Fatty liver disease. PPAR Res 2012; mice. Gastroenterology 2010; 139: 323-334.
2012: 912351. [50] Das K, Lin Y, Widen E, Zhang Y, Scherer PE.
[41] Foster FM, Traer CJ, Abraham SM, Fry MJ. The Chromosomal localization, expression pattern,
phosphoinositide (PI) 3-kinase family. J Cell Sci and promoter analysis of the mouse gene en-
2003; 116: 3037-3040. coding adipocyte-specific secretory protein
[42] Yuan J, Gao H, Sui J, Duan H, Chen WN, Ching Acrp30. Biochem Biophys Res Commun 2001;
CB. Cytotoxicity evaluation of oxidized single- 280: 1120-1129.
walled carbon nanotubes and graphene oxide [51] Hegarty BD, Turner N, Cooney GJ, Kraegen EW.
on human hepatoma HepG2 cells: an iTRAQ- Insulin resistance and fuel homeostasis: the
coupled 2D LC-MS/MS proteome analysis. role of AMP-activated protein kinase. Acta
Toxicol Sci 2012; 126: 149-161. Physiol (Oxf) 2009; 196: 129-145.
[43] Li X, Monks B, Ge Q, Birnbaum MJ. Akt/PKB [52] Sheng CH, Di J, Jin Y, Zhang YC, Wu M, Sun Y,
regulates hepatic metabolism by directly inhib- Zhang GZ. Resistin is expressed in human he-
iting PGC-1alpha transcription coactivator. Na- patocytes and induces insulin resistance. En-
ture 2007; 447: 1012-1016. docrine 2008; 33: 135-143.
[44] Liu Y, Han X, Bian Z, Peng Y, You Z, Wang Q,
Chen X, Qiu D, Ma X. Activation of liver X recep-
tors attenuates endotoxin-induced liver injury
in mice with nonalcoholic fatty liver disease.
Dig Dis Sci 2012; 57: 390-398.

1631 Int J Clin Exp Med 2014;7(7):1624-1631

You might also like