ArticlePDF AvailableLiterature Review

Nonalcoholic fatty liver disease: Molecular mechanisms for the hepatic steatosis

Authors:

Abstract and Figures

Liver plays a central role in the biogenesis of major metabolites including glucose, fatty acids, and cholesterol. Increased incidence of obesity in the modern society promotes insulin resistance in the peripheral tissues in humans, and could cause severe metabolic disorders by inducing accumulation of lipid in the liver, resulting in the progression of non-alcoholic fatty liver disease (NAFLD). NAFLD, which is characterized by increased fat depots in the liver, could precede more severe diseases such as non-alcoholic steatohepatitis (NASH), cirrhosis, and in some cases hepatocellular carcinoma. Accumulation of lipid in the liver can be traced by increased uptake of free fatty acids into the liver, impaired fatty acid beta oxidation, or the increased incidence of de novo lipogenesis. In this review, I would like to focus on the roles of individual pathways that contribute to the hepatic steatosis as a precursor for the NAFLD.
Content may be subject to copyright.
Copyri ght © 2013 by The Kore an Assoc iation f or the Stu dy of the Li ver
This i s an Open Acces s ar ticle distr ibuted under the t erms of the Creative Com mons A ttrib ution Non- Comme rcial Licen se (ht tp://creativecom mons.or g/li censes /by-nc/3.0/ )
which pe rmits un restri cted non -com mercia l use, dis tribut ion, and r eprodu ction i n any mediu m, provid ed the or iginal wo rk is pro perly ci ted.
pISSN 2287-2728
eISSN 2287-285X
http://dx.doi.org/10.3350/cmh.2013.19.3.210
Clinical and Molecular Hepatology 2013;19:210-215
Review
INTRODUCTION
In mammals, liver is a central organ for controlling the metabol-
ic homeostasis for carbohydrates, lipid, and proteins. Dysregula-
tion of liver functions could lead to the metabolic disorder that is
ultimately detrimental to the health of individuals. Recently, the
increased incidence of obesity is recognized as a major cause for
the promotion of metabolic diseases including non-alcoholic fatty
liver diseases (NAFLD), which is not only linked with other meta-
bolic diseases such as diabetes, but also invoke more severe liver
diseases including non-alcoholic steatohepatitis (NASH), hepatic
cirrhosis, and hepatocellular carcinoma (HCC).1
NAFLD can be characterized by the increased accumulation of
lipid in the liver, which can be stemmed from the multiple factors.
Increased lipolysis from the fat cells or the increased intake of di-
etary fat, followed by the enhancement of free fatty acids (FFA)
can explain this phenomenon.2 Mitochondrial dysfunction that is
associated with insulin resistance, which normally precedes the
NAFLD, could also cause lipid accumulation by impairment of fatty
acid beta oxidation.3 In addition, de novo lipogenesis in the liver
contributes greatly to the hepatic steatosis.4 Finally, reduction in
lipid clearance that is often associated with insulin resistance can
also exacerbate the condition (Fig. 1).5
Accumulation of lipid in the liver can further stimulate existing
Nonalcoholic fatty liver disease: molecular mechanisms
for the hepatic steatosis
Seung-Hoi Koo
Department of Life Sciences, Korea University, Seoul, Korea
Corresponding author : Seung-Hoi Koo
Department of Life Sciences, Korea University, 145 Anam-ro, Seongbuk-
gu, Seoul 136-713, Korea
Tel. +82-2-3290-3403, Fax. +82-2-3290-4144
E-mail; koohoi@korea.ac.kr
Abbreviations:
ACC, acetyl-CoA carboxy lase; ACOX, acyl- CoA oxidase; AMPK, AMP-activated
protein kinase; ap oB, apolipoprotein B; ChREBP, carbohydrate resp onse element
binding protein; CPT1, carnitine palmitoylt ransferase 1; DAG, diacylglycerol; DGAT,
acyl- CoA: diacylglycerol ac yltransferase; ELOVL6, long chain fatty acid elongase 6;
ER stress, endop lasmic reticulum stress; FABP, fatty a cid binding protein; FAS, fatty
acid synthas e; FAT, fatty aci d translocase; FATP, fatty acid tra nsporter protein; FFA,
free fatty acids; GPAT, glycerol-3-phosphate ac yltransferase; HCC, hepatocellular
carcinoma; HSL, hormone-s ensitive lipase; LXR alpha, live r X receptor alpha;
mTOR, mammalian target of r apamycin; NAFLD, non-alcoholic fat ty liver disease;
NASH, non-alco holic steatohepatitis; PGC-1, PPAR gamma co-activator 1; PKA,
protein kinase A; PPAR, peroxisome proliferator-activated receptor; SCD, stearoyl-
CoA desaturase; SIK s, salt inducible kinas es; SREBP-1c, sterol regulatory element
binding protein 1c; TG, trigl ycerides; VLDL, very low de nsity lipoprotein Received :
Jul. 31, 2013 /
Accepted :
Aug. 6, 2013
Liver plays a central role in the biogenesis of major metabolites including glucose, fatty acids, and cholesterol.
Increased incidence of obesity in the modern society promotes insulin resistance in the peripheral tissues in humans,
and could cause severe metabolic disorders by inducing accumulation of lipid in the liver, resulting in the progression
of non-alcoholic fatty liver disease (NAFLD). NAFLD, which is characterized by increased fat depots in the liver, could
precede more severe diseases such as non-alcoholic steatohepatitis (NASH), cirrhosis, and in some cases hepatocellular
carcinoma. Accumulation of lipid in the liver can be traced by increased uptake of free fatty acids into the liver, impaired
fatty acid beta oxidation, or the increased incidence of de novo lipogenesis. In this review, I would like to focus on the
roles of individual pathways that contribute to the hepatic steatosis as a precursor for the NAFLD. (Clin Mol Hepatol
2013;19:210-215)
Keywords: Free fatty acids; De novo lipogenesis; Fatty acid beta oxidation; TG secretion
211
Seung-Hoi Koo.
Nonalcoholic fatty liver disease: molecular mechanisms for the hepatic steatosis
http://www.e-cmh.org http://dx.doi.org/10.3350/cmh.2013.19.3.210
hepatic insulin resistance by generation of lipid-derived second
messengers such as diacylglycerol (DAG) and ceramides.6 Further-
more, lipid accumulation in the liver is also linked with the pro-
gression of endoplasmic reticulum stress (ER stress), mitochondria
stress, and impaired autophagy, resulting in the condition known
as lipotoxicity.7 This latter event can cause the immune response
in the Kupffer cells and hepatic stellate cells, which leads to the
progression of NASH, hepatic cirrhosis, and in some severe cases,
hepatocellular carcinoma.
In this review, we would like to delineate the molecular mecha-
nism for lipid accumulation in the liver as a major precursor for the
NAFLD. In particular, we will delineate the individual mechanisms
for the triglycerides (TG) synthesis and clearance that is critical in
mediating lipid homeostasis in the liver both under physiological
conditions and pathological conditions. Understanding of the mo-
lecular basis of these pathways could shed the insight into the po-
tential therapeutics in the treatment of this disease.
Fatty acids uptake
Free fatty acids (FFA) in the plasma can be taken up by the liver,
and serve as important sources for the TG synthesis in the liver.
Normally, plasma FFA is generated by white adipocytes via lipoly-
sis, which is induced by beta adrenergic receptor agonists such as
catecholamine under fasting conditions.8 This process involves the
regulation of protein kinase A (PK A)-dependent phosphorylation
and activation of hormone-sensitive lipase (HSL), a key rate limit-
ing enzyme in the lipolysis, to promote this pathway. This pathway
is reversed by insulin under feeding conditions, limiting the libera-
tion of FFA and rather inducing de novo lipogenesis in this tissue.
Upon insulin resistance that is associated with obesity, lipolysis is
hyperactivated in adipocytes, resulting in the increases in plasma
FFA.9 In addition, since obesity is often associated with increased
uptake of nutrition rich in lipid, we would expect to observe high-
er levels of precursors for TG synthesis in the liver.
The main plasma membrane transporters for FFA are fatty acid
transporter protein (FATP), caveolins, fatty acid translocase (FAT)/
CD36, and fatty acid binding protein (FABP). In mammals 6 mem-
bers of FATPs are found that contain a common motif for fatty
acid uptake and fatty acyl-CoA synthetase function.10 Among fam-
ily members, FATP2 and FATP5 are highly expressed in the liver,
and utilized as major FATPs for the normal physiological context.
Indeed, FATP5 knockout mice showed resistance to diet-induced
obesity and hepatic TG accumulation, although no clinical evi-
dence for the involvement of this FATP isoform in human obesity.11
Caveolins consist of three protein family members termed caveo-
lins 1, 2, and 3, and are found in the membrane structures called
caveolae, which are important for protein trafficking and the for-
mation of lipid droplets.12 Caveolin-1 knockout mice exhibited
lower TG accumulation in the liver and showed resistance to diet-
induced obesity, showing the importance of this protein in the TG
synthesis under obesity. FAT/CD36 is a transmembrane protein
that accelerates FA uptake via facilitated diffusion.13 Normally, this
protein is not highly expressed in the liver, but is enhanced by diet-
induced obesity. The hepatic expression of CD36 was positively
correlated with hepatic TG contents in NAFLD patients, underscor-
ing the potential importance of this transporter with this disease.14
FABPs are cytosolic lipid binding proteins that facilitate intracellu-
lar transport of FFAs.15 Among 9 isoforms, FABP1 and FABP5 are
highly expressed in the liver. Mice with liver-specific deletion of
FABP1 displayed resistance to diet-induced obesity, although he-
patic TG accumulation did not differ from that of wild type mice,
suggesting that contributions from other FABPs might be critical in
hepatic TG accumulation in this state.16 Indeed, expression of
FABP4 and FABP5 in the liver was correlated with hepatic fatty in-
filtration in NAFLD patients.17 Further study is necessary to inte-
grate roles of these fatty acid transporters in the hepatic FFA up-
take under the physiological conditions and the pathological
conditions.
Figure 1. Model for the TG accumulation in the liver in the early stage
of NAFLD. Hepatic steatosis can be stimulated via increased fatty acid
uptake, increased de novo lipogenesis, and decreased fatt y acid
oxidation followed by esterification for the TG synthesis. Additionally,
de creas es in VL DL secr eti on can also con tribut e to the lip id
accumulation in the liver. See texts for the molecular mechanisms in
detail.
Reduced
FA
oxidation
Hepatic TG
accumulation
Increased
FA
uptake (obesity)
Reduced
VLDL
secretion
De no vo
lipogenesis
212
Clin Mol Hepatol
Volume_
19 Number_3 September 2013
http://www.e-cmh.org
http://dx.doi.org/10.3350/cmh.2013.19.3.210
De novo lipogenesis
De novo lipogenesis is an integrated metabolic pathway that
comprises of glycolysis (conversion of glucose to acetyl-CoA), bio-
synthesis of saturated fatty acid followed by desaturation, and the
formation of TG. Key rate limiting enzymes in the process include
glucokinase and liver-type pyruvate kinase in the glycolysis, acetyl-
CoA carboxylase (ACC) and fatty acid synthase (FAS) in the fatty
acid synthesis, long chain fatt y acid elongase 6 (ELOVL6) and
stearoyl-CoA desaturase (SCD) in the formation of monounsatu-
rated fatty acids, and glycerol-3-phosphate acyltransferase (GPAT),
lipins, and acyl-CoA: diacylglycerol acyltransferase (DGAT) in the
formation of TG.18
FAS is a rate-limiting enzyme in the fatty acid biosynthesis and
catalyzes the last step in this pathway.19 Liver-specific knockout of
FAS in mice, however, displayed fatty liver phenotypes upon high
carbohydrate diet, perhaps due to the increase in hepatic malonyl-
CoA contents that would inhibit fatty acid beta oxidation.20 ACC
not only catalyzes a key rate-limiting step in the fatty acid biosyn-
thesis, but also is involved in the control of fatty acid oxidation by
synthesis of malonyl-CoA, an inhibitor for carnitine palmitoyl
transferase 1 (CPT1).21 Indeed, inhibition of liver-specific isoform
ACC1 in mice reduced hepatic TG levels in mice, by simultaneously
inhibiting fatty acid biosynthesis and activating fatty acid beta oxi-
dation in the liver.19 SCD1 is a microsomal enzyme that catalyses
the formation of monounsaturated long-chain fatty acids from
saturated fatty acyl-CoAs (E.g. conversion of palmitoyl-CoA (16:0)
to palmitoleoyl-CoA (16:1 n-7), and stearoyl-CoA (18:0) to oleoyl-
CoA (18:1 n-9)), and is predominantly expressed in the liver.22 De-
pletion of SCD1 in mice resulted in the reversal of hepatic steatosis
under western diet due to the decreased lipogenesis and increased
fatty acid beta oxidation.23 DGAT catalyzes the final step of TG
synthesis by catalyzing the acylation of diacylglycerol (DAG).24 In-
activation of hepatic isoform DGAT2 in obese mice resulted in the
significant reduction in hepatic TG contents, showing the in vivo
evidence for the importance of this protein in the de novo lipogen-
esis.25
Two major transcription factors for lipogenesis, sterol regulatory
element binding protein 1c (SREBP-1c) and carbohydrate response
element binding protein (ChREBP), are involved in the transcrip-
tional activation of genes encoding aforementioned rate-limiting
enzymes in the lipogenesis, and have been associated with in-
creased de novo lipogenesis in NAFLD.4 SREBP-1c is a member of
SREBP family that control transcriptional regulation of lipid metab-
olism.26 As ER-bound precursors, full-length SREBPs reside in the
ER by using its transmembrane domain in the middle. Transport of
SREBPs from the ER to the Golgi apparatus is mediated in part by
nutrient sensors SCAP and Insig, and the mature form of SREBPs
is generated by two consecutive proteolytic cleavages. The expres-
sion of SREBP-1c is highly induced by insulin under feeding condi-
tions and by saturated fatty acids upon high fat diet feeding in
mice, and the liver X receptor alpha (LXR alpha) is shown to be
critical in the transcriptional activation of SREBP-1c in this pro-
cess.27 The activity of SREBP-1c can be further activated by mam-
malian target of rapamycin (mTOR) pathway, while it can be inhib-
ited by PKA, AMP-activated protein kinase (AMPK), and salt
inducible kinases (SIKs).28-30 ChREBP was first identified as a regu-
lator for the hepatic glycolysis by activating transcription of L-PK
gene, and was later shown to be involved in the regulation of oth-
er lipogenic genes in the pathway.31 At low glucose conditions,
ChREBP is present in the cytosol by PK A-mediated phosphoryla-
tion, but undergoes dephosphorylation, association with its het-
erodimeric partner Mlx, and nuclear localization at high glucose
conditions, resulting in the transcriptional activation of target
genes in the liver. The additional role of AMPK in the regulation of
ChREBP was also suggested, although the exact mechanism is still
verified in vivo yet. In obese, insulin-resistant ob/ob mice, both
SREBP-1c and ChREBP are highly abundant in the liver, and reduc-
tion of either factor was shown to be beneficial in relieving hepatic
steatosis in mice, underscoring the importance of these lipogenic
transcription factors in de novo lipogenesis and the TG accumula-
tion in the liver.32,33
Fatty acid oxidative pathways
Fatty acid beta oxidation in mitochondria is a process to shorten
the fatty acids into acetyl-CoA, which can be later converted into
ketone bodies (beta hydroxybutyrate or acetoacetate) or can be
incorporated into the TCA cycle for the full oxidation.34 To initiate
the process, fatty acyl-CoAs should be transported across the mi-
tochondrial membranes by activity of a couple of CPTs. Fatty acyl-
CoAs are converted to fatty acyl-carnitines by CPT1 in the mito-
ch ondri al o uter mem brane for the transloc a tion int o the
intermembrane space. Fatty acyl-carnitines are then transported
across the mitochondrial outer membrane by carnitine acylcarni-
tine translocase. CPT2 converts fatty acyl-carnitines to fatty acyl-
CoAs for the fatty acid beta oxidation inside the mitochondrial
matrix. The first step involves the beta dehydrogenation of the ac-
yl-CoA ester by chain length-specific acyl-CoA dehydrogenases
(e.g. VLCAD, LCAD, and MCAD). Indeed, mice deficient in MCAD
213
Seung-Hoi Koo.
Nonalcoholic fatty liver disease: molecular mechanisms for the hepatic steatosis
http://www.e-cmh.org http://dx.doi.org/10.3350/cmh.2013.19.3.210
and VLCAD develop hepatic steatosis, supporting the role of these
proteins and the fatty acid beta oxidation in the hepatic TG con-
tent.35 2-enoyl-CoA hydratase, 3-hydroxyacyl-CoA dehydrogenase
and 3-oxoacyl-CoA thiolase are subsequently involved in the fatty
acid beta oxidation process to complete the conversion of acyl-
CoA ester into acetyl-CoA.
Under fasting conditions, fatty acid beta oxidation is enhanced
in part via inactivation of ACC, resulting in the reduced production
of malonyl-CoA that serves as a potent inhibitor of CPT1.21 Chronic
starvation also increases expression of beta oxidation genes via
transcriptional mechanisms. Peroxisome proliferator-activated re-
ceptor (PPAR) alpha and its co-activator PPAR gamma co-activator
1 (PGC-1) alpha are critical in enhancing expression of target
genes including CPT1, LCAD, MCAD, and acyl-CoA oxidase
(ACOX).36 Starvation-dependent activation of AMPK and sirtuins
could also enhance expression of these genes by directly modify-
ing and activating PGC-1 alpha.37,38 The clinical implication of im-
paired mitochondrial beta oxidation on the progression of NAFLD
is not conclusive, and contradicting reports have been published.
Further study is necessary to delineate the role of fatty acid beta
oxidation on hepatic lipid accumulation and the progression of
NAFLD.39
TG secretion
In the liver, TG secretion is achieved via the formation of very
low density lipoprotein (VLDL).40 VLDL consists of hydrophobic
core lipids containing TGs and cholesterol esters, which is covered
by hydrophilic phospholipids and apolipoprotein B (apoB) 100.
ApoB 100 is a liver-specific ApoB that is critical in the VLDL as-
sembly, while apoB 48 in the intestine is associated with chylomi-
cron formation. The VLDL assembly process occurs initially in the
rough ER during the translation and translocation of the apoB
100, resulting in the formation of a partially lipidated apoB 100.
The partially lipidated apoB 100, termed the pre-VLDL is then
transported into the Golgi for the maturation, and subsequently
released from the liver via exocytosis. Indeed, hepatic steatosis
was reported in patients carrying mutations in apoB 100 (hypo-
betalipoproteinemia) and in MTP (abetalipoproteinemia), under-
scoring the importance of these proteins in the lipid homeostasis
in humans.41,42
Anabolic hormone insulin is critical in the regulation of VLDL as-
sembly and secretion. Insulin plays a role in the degradation of
apoB 100, perhaps utilizing an autophagy-dependent pathway.43
Furthermore, insulin inhibits the transcription of MTP, by phospha-
tidylinositol 3 kinase/Akt-dependent regulation. Akt phosphory-
lates and inhibits transcription factor FoxA2, a critical forkhead
box factor for activating expression of MTP at the transcription
level.44,45 Upon insulin resistance, perturbation of this process re-
sults in hypertriglyceridemia via increased TG secretion. However,
the rate of TG secretion cannot keep up with the increased rate of
TG synthesis in this condition, resulting in the hepatic steatosis in
spite of the increased VLDL secretion. Similar phenotype is ob-
served in NAFLD patients, which exhibit both hypertriglyceridemia
and hepatic steatosis.4 6 Prolonged exposure of the liver to FFA
would promotes ER stress and other oxidative stress in the liver,
leading to the degradation of apoB 100, decrease in the VLDL se-
cretion, and worsening of hepatic steatosis.
CONCLUSIONS
The balance between the TG uptake/synthesis and TG hydroly-
sis/secretion is critical in the maintenance of lipid homeostasis in
the liver. Clearly, perturbation of any of these pathways could be
detrimental to the lipid metabolism. In the case of NAFLD, the
progression of hepatic steatosis can stem from the increased FFA
uptake and de novo lipogenesis for the increased TG synthesis,
and the decreased TG hydrolysis and fatty acid beta oxidation. Re-
duced TG secretion via VLDL could also promote hepatic lipid ac-
cumulation, although the VLDL secretion was rather increased in
NAFLD patients. Understanding of molecular mechanisms of each
pathway is critical in pursuing the development of therapeutics of
NAFLD in the future.
Acknowledgements
This work was supported by the National Research Foundation
of Ko r ea (g r an t nos . : NR F -2010 - 0015 09 8 an d N R F-
2012M3A9B6055345), funded by the Ministry of Science, ICT &
Future Planning, Republic of Korea, and a grant of the Korea
Health technology R&D Project (grant no : A111345), Ministry of
Health & Welfare, Korea.
Conicts of Interest
The authors have no conflicts to disclose.
214
Clin Mol Hepatol
Volume_
19 Number_3 September 2013
http://www.e-cmh.org
http://dx.doi.org/10.3350/cmh.2013.19.3.210
REFERENCES
1. Browning JD, Szczepaniak LS, Dobbins R, Nuremberg P, Horton
JD, Cohen JC, et al. Prevalence of hepatic steatosis in an urban
population in the United States: impact of ethnicity. Hepatology
2004;40:1387-1395.
2. Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD,
Park s EJ. Sources of fatty acids stored in liver and secreted via
lipoproteins in patients with nonalcoholic fatty liver disease. J Clin
Invest 2005;115:1343-1351.
3. Fromenty B, Robin MA, Igoudjil A, Mansouri A, Pessayre D. The ins
and outs of mitochondrial dysfunc tion in NASH. Diabetes Metab
2004;30:121-138.
4. Postic C, Girard J. Contribution of de novo fatty acid synthesis to
hepatic steatosis and insulin resistance: lessons from genetically
engineered mice. J Clin Invest 2008;118:829-838.
5. Fabbrini E, Mohammed BS, Magkos F, Korenblat KM, Patterson
BW, Klein S. Alterations in adipose tissue and hepatic lipid kinet-
ics in obese men and women with nonalcoholic fatty liver disease.
Gastroenterology 2008;134:424-431.
6. Jor nay vaz FR, Sh ulman GI. Dia c ylglycer ol act ivation of pro -
tein kinase Cepsilon and hepatic insulin resistance. Cell Metab
2012;15:574-584.
7. Neuschwander-Tetri BA. Hepatic lipotoxicity and the pathogenesis
of nonalcoholic steatohepatitis: the central role of nontriglyceride
fatty acid metabolites. Hepatology 2010;52:774-788.
8. Arner P. Human fat cell lipolysis: biochemistry, regulation and clini-
cal role. Best Pract Res Clin Endocrinol Metab 2005;19:471-482.
9. Delarue J, Magnan C. Free fatty acids and insulin resistance. Curr
Opin Clin Nutr Metab Care 2007;10:142-148.
10. Doege H, Stahl A. Protein-mediated fat ty acid uptake: novel in-
sight s from in vivo models. Physiology (Bethesda) 2006;21:259-
268.
11. Doege H, Baillie RA, Or tegon AM, Tsang B, Wu Q, Punreddy S, et
al. Targeted deletion of FATP5 reveals multiple functions in liver
metabolism: alterations in hepatic lipid homeostasis. Gastroenterol-
ogy 2006;130:1245-1258.
12. Fernandez MA, Albor C, Ingelmo-Torres M, Nixon SJ, Ferguson C,
Kurzchalia T, et al. Caveolin-1 is essential for liver regeneration. Sci-
ence 2006;313:1628-1632.
13. Silverstein RL, Febbraio M. CD36, a scavenger receptor involved
in immunity, metabolism, angiogenesis, and behavior. Sci Signal
2009;2:re3.
14. Greco D, Kotronen A, Westerbacka J, Puig O, Arkkila P, Kiviluoto T,
et al. Gene expression in human NAFLD. Am J Physiol Gastrointest
Liver Physiol 2008;294:G1281- G1287.
15. Makowski L, Hotamisligil GS. The role of fatty acid binding pro-
teins in metabolic syndrome and atherosclerosis. Curr Opin Lipidol
2005;16:543-548.
16. Newberry EP, Xie Y, Kennedy S, Han X, Buhman KK, Luo J, et al.
Decreased hepatic triglyceride accumulation and altered fatty acid
uptake in mice with deletion of the liver fatty acid-binding protein
gene. J Biol Chem 2003;278:51664-51672.
17. Westerbacka J, Kolak M, Kiviluoto T, Arkkila P, Siren J, Hamsten
A, et al. Genes involved in fatty acid partitioning and binding, li-
polysis, monocyte/macrophage recruitment, and inflammation are
overexpressed in the human fatty liver of insulin-resistant subjects.
Diabetes 2007;56:2759-2765.
18. Towle HC, Kaytor EN, Shih HM. Regulation of the expression of lipo-
genic enzyme genes by carbohydrate. Annu Rev Nutr 1997;17:405-
433.
19. Abu-Elheiga L, Matzuk MM, Kordari P, Oh W, Shaikenov T, Gu Z.
Mutant mice lacking acetyl-CoA carboxylase 1 are embryonically
lethal. Proc Natl Acad Sci U S A 2005;102:12011-12016.
20. Chakravarthy MV, Pan Z, Zhu Y, Tordjman K, Schneider JG, Cole-
man T, et al. “New” hepatic fat activates PPARalpha to maintain
glucose, lipid, and cholesterol homeostasis. Cell Metab 2005;1:309-
322.
21. Munday MR. Regulation of mammalian acetyl-CoA carboxylase.
Biochem Soc Trans 2002;30:1059-1064.
22. Miyazaki M, Kim YC, Ntambi JM. A lipogenic diet in mice with a
disruption of the stearoyl-CoA desaturase 1 gene reveals a strin-
gent requirement of endogenous monounsaturated fatty acids for
triglyceride synthesis. J Lipid Res 2001;42:1018-1024.
23. Jiang G, Li Z, Liu F, Ellsworth K, Dallas-Yang Q, Wu M, et al. Pre-
vention of obesity in mice by antisense oligonucleotide inhibitors of
stearoyl-CoA desaturase-1. J Clin Invest 2005;115:1030-1038.
24. Cases S, Smith SJ, Zheng YW, Myers HM, Lear SR, Sande E, et al.
Identification of a gene encoding an acyl CoA:diacylglycerol ac-
yltransferase, a key enzyme in triacylglycerol synthesis. Proc Natl
Acad Sci U S A 1998;95:13018-13023.
25. Yu XX, Murray SF, Pandey SK, Booten SL, Bao D, Song XZ, et al.
Antisense oligonucleotide reduction of DGAT2 expression improves
hepatic steatosis and hyperlipidemia in obese mice. Hepatology
2005;42:362-371.
26. Horton JD, Goldstein JL, Brown MS. SREBPs: activators of the com-
plete program of cholesterol and fatt y acid synthesis in the liver. J
Clin Invest 2002;109:1125-1131.
27. Chen G, Liang G, Ou J, Goldstein JL, Brown MS. Central role for
liver X receptor in insulin-mediated activation of Srebp-1c transcrip-
tion and stimulation of fatty acid synthesis in liver. Proc Natl Acad
Sci U S A 2004;101:11245-11250.
28. Porstmann T, Santos CR, Griffiths B, Cully M, Wu M, Leevers S, et
al. SREBP activity is regulated by mTORC1 and contributes to Akt-
dependent cell growth. Cell Metab 2008;8:224-236.
29. Lu M, Shyy JY. Sterol regulatory element-binding protein 1 is nega-
215
Seung-Hoi Koo.
Nonalcoholic fatty liver disease: molecular mechanisms for the hepatic steatosis
http://www.e-cmh.org http://dx.doi.org/10.3350/cmh.2013.19.3.210
tively modulated by PKA phosphorylation. Am J Physiol Cell Physiol
2006;290:C1477- C1486.
30. Yoon YS, Seo WY, Lee MW, Kim ST, Koo SH. Salt-inducible kinase
regulates hepatic lipogenesis by controlling SREBP-1c phosphoryla-
tion. J Biol Chem 2009;284:10446-10452.
31. Uyeda K, Repa JJ. Carbohydrate response element binding protein,
ChREBP, a transcription factor coupling hepatic glucose utilization
and lipid synthesis. Cell Metab 2006;4:107-110.
32. Dentin R, Benhamed F, Hainault I, Fauveau V, Foufelle F, Dyck JR,
et al. Liver-specific inhibition of ChREBP improves hepatic steatosis
and insulin resistance in ob/ob mice. Diabetes 2006;55:2159-2170.
33. Shimomura I, Bashmakov Y, Horton JD. Increased levels of nuclear
SREBP-1c associated with fatty livers in two mouse models of dia-
betes mellitus. J Biol Chem 1999;274:30028-30032.
34. Eaton S, Bartlett K, Pourfarzam M. Mammalian mitochondrial beta-
oxidation. Biochem J 1996;320:345-357.
35. Tolwani RJ, Hamm DA, Tian L, Sharer JD, Vockley J, Rinaldo P, et al.
Medium-chain acyl-CoA dehydrogenase deficiency in gene-targeted
mice. PLoS Genet 2005;1:e23.
36. Reddy JK, Hashimoto T. Peroxisomal beta-oxidation and peroxisome
proliferator-activated receptor alpha: an adaptive metabolic system.
Annu Rev Nutr 2001;21:193-230.
37. Foster DW. Malonyl-CoA: the regulator of fatty acid synthesis and
oxidation. J Clin Invest 2012;122:1958-1959.
38. Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of me-
tabolism and healthspan. Nat Rev Mol Cell Biol 2012;13:225-238.
39. Samuel VT, Shulman GI. Mechanisms for insulin resistance: com-
mon threads and missing links. Cell 2012;148:852-871.
40. Hussain MM , Shi J, Dreizen P. Microsomal triglyceride transfer
protein and its role in apoB-lipoprotein as se mbly. J Lipid Res
2003;44:22-32.
41. Tanoli T, Yue P, Yablonskiy D, Schonfeld G. Fatty liver in familial hy-
pobetalipoproteinemia: roles of the APOB defects, intra-abdominal
adipose tissue, and insulin sensitivity. J Lipid Res 2004;45:941-947.
42. Berriot-Varoqueaux N, Aggerbeck LP, Samson-Bouma M, Wetterau
JR. The role of the microsomal triglygeride transfer protein in abet-
alipoproteinemia. Annu Rev Nutr 2000;20:663-697.
43. Ginsb erg HN, Fisher EA. Th e ever-expanding role of degrada-
tion in the regulation of apolipoprotein B metabolism. J Lipid Res
2009;50(Suppl):S162-S166.
44. Koo SH, Montminy M. Fatt y acids and insulin resistance: a perfect
storm. Mol Cell 2006;21:449-450.
45. Wolfrum C, Stoffel M. Coactivation of Foxa2 through Pgc-1beta
promotes liver fatty acid oxidation and triglyceride/VLDL secretion.
Cell Metab 2006;3:99-110.
46. Choi SH, Ginsberg HN. Increased very low density lipoprotein (VLDL)
secretion, hepatic steatosis, and insulin resistance. Trends Endocri-
nol Metab 2011;22:353-363.
... In this context, results found in these experiments indicate minor hepatotoxicity, microvesicular steatosis that can result in increased fat accumulation in liver cells, in all enzyme treated groups and an increased organ size, except for the S206C group. This may be caused by an impairment of liver metabolic functions that includes defective fatty acid oxidations, enhanced lipogenesis, irregular triglyceride secretion or increased absorptions of fat acids from the diet [38,39]. Hepatic failure, as platelet decrease, may be related with anti-asparaginase antibodies such as IgG and IgE, which also may form and accumulate immune complexes and could be responsible for the increased organ size on WT and P40S enzymes treated groups, unlike S206C exposed group. ...
Preprint
Full-text available
Acute lymphoblastic leukaemia is currently treated with bacterial L-Asparaginase; however, its side effects bring up the special need for improved and efficient novel enzymes development. Previously, we obtained low anti-asparaginase antibody production and high serum enzyme half-life in mice with the P40S/S206C mutant; however, its specific activity was significantly reduced. Thus, our aim was to test single mutants, S206C and P40S, for in vitro and in vivo assays. Our results showed that the drop in specific activity was caused by P40S substitution. Besides, our single mutants were highly stable in biological environment simulation, unlike double mutant P40S/S206C. The in vitro cell viability assay demonstrated that mutant enzymes have higher cytotoxic effect than WT on T-cells derived ALL and on some solid cancer cell lines. The in vivo assays were performed in mice to identify toxicological effects, to evoke immunological response and to study the enzymes pharmacokinetics. From these tests, none of the enzymes was toxic; however, S206C evoked lower physiological changes and immune responses. In relation to pharmacokinetic profile, S206C has two-fold higher activity than WT and P40S two hours after injection. In conclusion, we present bioengineered E. coli asparaginases with high specific enzyme activity and lesser side effects.
... In this context, the results of these experiments indicate minor hepatotoxicity and microvesicular steatosis, which can result in increased fat accumulation in liver cells in all enzyme-treated groups and increased organ size, except for the S206C group. This may be caused by an impairment of liver metabolic functions, including defective fatty acid oxidation, enhanced lipogenesis, irregular triglyceride secretion or increased absorptions of fatty acids from the diet [41,42]. Hepatic failure, like platelet decrease, may be related to anti-asparaginase antibodies such as IgG and IgE, which may also form and accumulate immune complexes and could be responsible for the increased organ size in WT-and P40S-enzyme-treated groups, unlike the S206C-exposed group. ...
Article
Full-text available
Acute lymphoblastic leukaemia is currently treated with bacterial L-asparaginase; however, its side effects raise the need for the development of improved and efficient novel enzymes. Previously, we obtained low anti-asparaginase antibody production and high serum enzyme half-life in mice treated with the P40S/S206C mutant; however, its specific activity was significantly reduced. Thus, our aim was to test single mutants, S206C and P40S, through in vitro and in vivo assays. Our results showed that the drop in specific activity was caused by P40S substitution. In addition, our single mutants were highly stable in biological environment simulation, unlike the double-mutant P40S/S206C. The in vitro cell viability assay demonstrated that mutant enzymes have a higher cytotoxic effect than WT on T-cell-derived ALL and on some solid cancer cell lines. The in vivo assays were performed in mice to identify toxicological effects, to evoke immunological responses and to study the enzymes’ pharmacokinetics. From these tests, none of the enzymes was toxic; however, S206C elicited lower physiological changes and immune/allergenic responses. In relation to the pharmacokinetic profile, S206C exhibited twofold higher activity than WT and P40S two hours after injection. In conclusion, we present bioengineered E. coli asparaginases with high specific enzyme activity and fewer side effects.
... The "double hit" process destroys the dynamic balance between lipid synthesis and lipid consumption, resulting in excessive accumulation of liver fat and, in turn, leading to liver steatosis. Therefore, accelerating lipid consumption and/or inhibiting lipid synthesis is considered an effective solution to reducing hepatic lipid accumulation [10]. ...
Article
Full-text available
Non-alcoholic fatty liver disease (NAFLD) is a clinicopathological syndrome characterized by diffuse hepatocellular steatosis due to fatty deposits in hepatocytes, excluding alcohol and other known liver injury factors. However, there are no specific drugs for the clinical treatment of NAFLD. Therefore, research on the pathogenesis of NAFLD at the cellular and molecular levels is a promising approach to finding therapeutic targets and developing targeted drugs for NAFLD. Pin1 is highly expressed during adipogenesis and contributes to adipose differentiation, but its specific mechanism of action in NAFLD is unclear. In this study, we investigated the role of Pin1 in promoting the development of NAFLD and its potential mechanisms in vitro and in vivo. First, Pin1 was verified in the NAFLD model in vitro using MCD diet-fed mice by Western Blot, RT-qPCR and immunohistochemistry (IHC) assays. In the in vitro study, we used the oleic acid (OA) stimulation-induced lipid accumulation model and examined the lipid accumulation in each group of cells by oil red O staining as well as BODIPY staining. The results showed that knockdown of Pin1 inhibited lipid accumulation in hepatocytes in an in vitro lipid accumulation model and improved lipid indices and liver injury levels. Moreover, in vivo, WT and Pin1-KO mice were fed a methionine-choline deficient (MCD) diet for 4 weeks to induce the NAFLD model. The effects of Pin1 on lipid accumulation, hepatic fibrosis, and oxidative stress were evaluated by biochemical analysis, glucose and insulin tolerance tests, histological analysis, IHC, RT-qPCR and Western blot assays. The results indicate that Pin1 knockdown significantly alleviated hepatic steatosis, fibrosis and inflammation in MCD-induced NAFLD mice, improved glucose tolerance and alleviated insulin resistance in mice. Further studies showed that the AMPK/ACC1 signalling pathway might take part in the process by which Pin1 regulates NAFLD, as evidenced by the inhibition of the AMPK/ACC1 pathway. In addition, immunofluorescence (IF), coimmunoprecipitation (Co-IP) and GST pull-down experiments also showed that Pin1 interacts directly with ACC1 and inhibits ACC1 phosphorylation levels. Our study suggests that Pin1 promotes NAFLD progression by inhibiting the activation of the AMPK/ACC1 signalling pathway, and it is possible that this effect is achieved by Pin1 interacting with ACC1 and inhibiting the phosphorylation of ACC1.
Article
Hepatic steatosis is the initial manifestation of abnormal liver functions and often leads to liver diseases such as nonalcoholic fatty liver disease in humans and fatty liver syndrome in animals. In this study, we conducted a comprehensive analysis of a large chicken population consisting of 705 adult hens by combining host genome resequencing; liver transcriptome, proteome, and metabolome analysis; and microbial 16S ribosomal RNA gene sequencing of each gut segment. The results showed the heritability (h2 = 0.25) and duodenal microbiability (m2 = 0.26) of hepatic steatosis were relatively high, indicating a large effect of host genetics and duodenal microbiota on chicken hepatic steatosis. Individuals with hepatic steatosis had low microbiota diversity and a decreased genetic potential to process triglyceride output from hepatocytes, fatty acid β-oxidation activity, and resistance to fatty acid peroxidation. Furthermore, we revealed a molecular network linking host genomic variants (GGA6: 5.59–5.69 Mb), hepatic gene/protein expression (PEMT, phosphatidyl-ethanolamine N-methyltransferase), metabolite abundances (folate, S-adenosylmethionine, homocysteine, phosphatidyl-ethanolamine, and phosphatidylcholine), and duodenal microbes (genus Lactobacillus) to hepatic steatosis, which could provide new insights into the regulatory mechanism of fatty liver development.
Article
Full-text available
In women who are getting older, the quantity and quality of their follicles or oocytes and decline. This is characterized by decreased ovarian reserve function (DOR), fewer remaining oocytes, and lower quality oocytes. As more women choose to delay childbirth, the decline in fertility associated with age has become a significant concern for modern women. The decline in oocyte quality is a key indicator of ovarian aging. Many studies suggest that age-related changes in oocyte energy metabolism may impact oocyte quality. Changes in oocyte energy metabolism affect adenosine 5'-triphosphate (ATP) production, but how related products and proteins influence oocyte quality remains largely unknown. This review focuses on oocyte metabolism in age-related ovarian aging and its potential impact on oocyte quality, as well as therapeutic strategies that may partially influence oocyte metabolism. This research aims to enhance our understanding of age-related changes in oocyte energy metabolism, and the identification of biomarkers and treatment methods.
Article
Chlorogenic acid (Chl), isochlorogenic acid A (Isochl A), and isochlorogenic acid B (Isochl B) are naturally occurring phenolic compounds, which have been shown to exert a regulatory effect on lipid metabolism. However, the mechanism underlying this effect remains unclear. Herein, we investigated the inhibitory effects and underlying mechanisms of these three phenolic compounds on oleic acid (OA)‐induced HepG2 cells and high‐fat diet (HFD)‐fed zebrafish. Lipid accumulation and triacylglycerol levels increased in OA‐induced cells, which was attenuated by Chl, Isochl A, and Isochl B. Moreover, the levels of malondialdehyde (MDA) and reactive oxygen species (ROS) decreased, while superoxide dismutase (SOD) levels increased by Chl, Isochl A and Isochl B treatment. Western blot analysis demonstrated that Chl, Isochl A and Isochl B reduced the expression of lipogenesis‐related protein, including fatty acid synthase (FAS), acetyl‐CoA carboxylase (ACC) and peroxisome proliferator‐activated receptor gamma (PPARγ). Moreover, peroxisome proliferator‐activated receptor alpha gamma (PPARα) was increased by Chl, Isochl A, and Isochl B treatment. In addition, our results indicated that Chl, Isochl A and Isochl B decreased lipid profiles and lipid accumulation in HFD‐fed zebrafish.
Article
Full-text available
Triacylglycerols are quantitatively the most important storage form of energy for eukaryotic cells. Acyl CoA:diacylglycerol acyltransferase (DGAT, EC 2.3.1.20) catalyzes the terminal and only committed step in triacylglycerol synthesis, by using diacylglycerol and fatty acyl CoA as substrates. DGAT plays a fundamental role in the metabolism of cellular diacylglycerol and is important in higher eukaryotes for physiologic processes involving triacylglycerol metabolism such as intestinal fat absorption, lipoprotein assembly, adipose tissue formation, and lactation. DGAT is an integral membrane protein that has never been purified to homogeneity, nor has its gene been cloned. We identified an expressed sequence tag clone that shared regions of similarity with acyl CoA:cholesterol acyltransferase, an enzyme that also uses fatty acyl CoA as a substrate. Expression of a mouse cDNA for this expressed sequence tag in insect cells resulted in high levels of DGAT activity in cell membranes. No other acyltransferase activity was detected when a variety of substrates, including cholesterol, were used as acyl acceptors. The gene was expressed in all tissues examined; during differentiation of NIH 3T3-L1 cells into adipocytes, its expression increased markedly in parallel with increases in DGAT activity. The identification of this cDNA encoding a DGAT will greatly facilitate studies of cellular glycerolipid metabolism and its regulation.
Article
Full-text available
Central and peripheral insulin-like peptides (ILPs), which include insulin, insulin-like growth factor 1 (IGF1) and IGF2, exert many effects in the brain. Through their actions on brain growth and differentiation, ILPs contribute to building circuitries that subserve metabolic and behavioural adaptation to internal and external cues of energy availability. In the adult brain each ILP has distinct effects, but together their actions ultimately regulate energy homeostasis - they affect nutrient sensing and regulate neuronal plasticity to modulate adaptive behaviours involved in food seeking, including high-level cognitive operations such as spatial memory. In essence, the multifaceted activity of ILPs in the brain may be viewed as a system organization involved in the control of energy allocation.
Article
Full-text available
CD36 is a membrane glycoprotein present on platelets, mononuclear phagocytes, adipocytes, hepatocytes, myocytes, and some epithelia. On microvascular endothelial cells, CD36 is a receptor for thrombospondin-1 and related proteins and functions as a negative regulator of angiogenesis. On phagocytes, through its functions as a scavenger receptor recognizing specific oxidized phospholipids and lipoproteins, CD36 participates in internalization of apoptotic cells, certain bacterial and fungal pathogens, and modified low-density lipoproteins, thus contributing to inflammatory responses and atherothrombotic diseases. CD36 also binds long-chain fatty acids and facilitates their transport into cells, thus participating in muscle lipid utilization, adipose energy storage, and gut fat absorption and possibly contributing to the pathogenesis of metabolic disorders, such as diabetes and obesity. On sensory cells, CD36 is involved in insect pheromone signaling and rodent fatty food preference. The signaling pathways downstream of CD36 involve ligand-dependent recruitment and activation of nonreceptor tyrosine kinases, specific mitogen-activated protein kinases, and the Vav family of guanine nucleotide exchange factors; modulation of focal adhesion constituents; and generation of intracellular reactive oxygen species. CD36 in many cells is localized in specialized cholesterol-rich membrane microdomains and may also interact with other membrane receptors, such as tetraspanins and integrins. Identification of the precise CD36 signaling pathways in specific cells elicited in response to specific ligands may yield novel targets for drug development.
Article
In the catabolic state with no food intake, the liver generates ketones by breaking down fatty acids. During the nocturnal fast or longer starvation periods, this protects the brain, which cannot oxidize fatty acids. In 1977, we published a study in the JCI noting the surprising realization that malonyl-CoA, the substrate of fatty acid synthesis, was also an inhibitor of fatty acid oxidation. Subsequent experiments have borne out this finding and furthered our understanding of molecular metabolism.
Article
Nonalcoholic fatty liver disease (NAFLD) is now the most frequent chronic liver disease in Western societies, affecting one in four adults in the USA, and is strongly associated with hepatic insulin resistance, a major risk factor in the pathogenesis of type 2 diabetes. Although the cellular mechanisms underlying this relationship are unknown, hepatic accumulation of diacylglycerol (DAG) in both animals and humans has been linked to hepatic insulin resistance. In this Perspective, we discuss the role of DAG activation of protein kinase Cε as the mechanism responsible for NAFLD-associated hepatic insulin resistance seen in obesity, type 2 diabetes, and lipodystrophy.
Article
Insulin resistance is a complex metabolic disorder that defies explanation by a single etiological pathway. Accumulation of ectopic lipid metabolites, activation of the unfolded protein response (UPR) pathway, and innate immune pathways have all been implicated in the pathogenesis of insulin resistance. However, these pathways are also closely linked to changes in fatty acid uptake, lipogenesis, and energy expenditure that can impact ectopic lipid deposition. Ultimately, these cellular changes may converge to promote the accumulation of specific lipid metabolites (diacylglycerols and/or ceramides) in liver and skeletal muscle, a common final pathway leading to impaired insulin signaling and insulin resistance.
Article
Since the beginning of the century, the mammalian sirtuin protein family (comprising SIRT1-SIRT7) has received much attention for its regulatory role, mainly in metabolism and ageing. Sirtuins act in different cellular compartments: they deacetylate histones and several transcriptional regulators in the nucleus, but also specific proteins in other cellular compartments, such as in the cytoplasm and in mitochondria. As a consequence, sirtuins regulate fat and glucose metabolism in response to physiological changes in energy levels, thereby acting as crucial regulators of the network that controls energy homeostasis and as such determines healthspan.
Article
Insulin resistance (IR) affects not only the regulation of carbohydrate metabolism but all aspects of lipid and lipoprotein metabolism. IR is associated with increased secretion of VLDL and increased plasma triglycerides, as well as with hepatic steatosis, despite the increased VLDL secretion. Here we link IR with increased VLDL secretion and hepatic steatosis at both the physiologic and molecular levels. Increased VLDL secretion, together with the downstream effects on high density lipoprotein (HDL) cholesterol and low density lipoprotein (LDL) size, is proatherogenic. Hepatic steatosis is a risk factor for steatohepatitis and cirrhosis. Understanding the complex inter-relationships between IR and these abnormalities of liver lipid homeostasis will provide insights relevant to new therapies for these increasing clinical problems.
Article
A significant body of evidence now forces us to rethink the causes of NASH. Once thought to be a disease caused by triglyceride accumulation in hepatocytes with subsequent oxidant stress and lipid peroxidation causing inflammation and fibrosis, new data from animal studies and a limited number of human studies now provide convincing evidence that triglyceride accumulation does not cause insulin resistance or cellular injury in the liver. The lipotoxic liver injury hypothesis for the pathogenesis of NASH suggests that we need to focus our therapeutic efforts on reducing the burden of fatty acids going to the liver or being synthesized in the liver. This can be accomplished by improving insulin sensitivity at the level of adipose tissue to prevent inappropriate peripheral lipolysis and by preventing unnecessary de novo lipogenesis in the liver. Excess carbohydrates are the major substrates for de novo lipogenesis, and thus, reducing carbohydrate consumption through dietary changes and increasing muscle glucose uptake through exercise remain important cornerstones of treatment and prevention of lipotoxic liver injury, a disease hitherto called NASH.