Abstract
Significance: Fatty liver disease is a major liver disorder in the modern societies. Comprehensive understanding of the pathophysiology and molecular mechanisms is essential for the prevention and treatment of the disease.
Recent Advances: Remarkable progress has been made in the recent years in basic and translational research in the field of fatty liver disease. Multiple signaling pathways have been implicated in the development of fatty liver disease, including AMP-activated protein kinase, mechanistic target of rapamycin kinase, endoplasmic reticulum stress, oxidative stress, inflammation, transforming growth factor β, and yes1-associated transcriptional regulator/transcriptional coactivator with PDZ-binding motif (YAP/TAZ). In addition, critical molecular regulations at the transcriptional and epigenetic levels have been linked to the pathogenesis of fatty liver disease.
Critical Issues: Some critical issues remain to be solved so that research findings can be translated into clinical applications. Robust and reliable biomarkers are needed for diagnosis of different stages of the fatty liver disease. Effective and safe molecular targets remain to be identified and validated. Prevention strategies require solid scientific evidence and population-wide feasibility.
Future Directions: As more data are generated with time, integrative approaches are needed to comprehensively understand the disease pathophysiology and mechanisms at multiple levels from population, organismal system, organ/tissue, to cell. The interactions between genes and environmental factors require deeper investigation for the purposes of prevention and personalized treatment of fatty liver disease. Antioxid. Redox Signal. 35, 689–717.
Keywords: signaling, epigenetic, transcription factor, steatohepatitis, fibrosis
Introduction
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease in the world, estimated to be ∼25% of the global population (29). NAFLD manifests a spectrum of hepatic disorders from simple steatosis to nonalcoholic steatohepatitis (NASH), and in some cases progresses to liver cirrhosis and cancer (29). Obesity, overnutrition, and physical inactivity are common risk factors for NAFLD (29). In addition, alcohol-related liver disease (ALD) is a common hepatic disorder in chronic alcohol drinkers (6). ALD shares similar progressive characteristics with NAFLD. In this review, we aimed to summarize recent developments in both ALD and NAFLD with a focus on signal transduction and epigenetic and transcriptional regulation.
Signal Transduction in Fatty Liver Disease
AMP-activated protein kinase signaling
AMP-activated protein kinase (AMPK) senses low energy status and plays a critical role in hepatic metabolic homeostasis (Fig. 1). AMPK is composed of three subunits: catalytic α subunit and regulatory β and γ subunits. AMPK activity is stimulated by nutrient deprivation and inhibited by overnutrition and alcohol consumption (56). Multiple factors can activate AMPK, including liver kinase B1 (LKB1), TGFβ (transforming growth factor β)-activated kinase 1 (TAK1), calcium/calmodulin-dependent protein kinase kinase β (CAMKKβ), and sestrins (SESNs) (56).
FIG. 1.
AMPK signaling and function in fatty live disease. AMPK activity is regulated by cellular energetic and nutritional status. As a response, AMPK controls multiple cellular processes, including hepatic lipid metabolism, oxidative stress, inflammation, fibrosis, and cell death. ACC1/2, acetyl-CoA carboxylases 1 and 2; AMPK, AMP-activated protein kinase; CAMKKβ, calcium/calmodulin-dependent protein kinase kinase beta; Casp6, caspase-6; CPT1, carnitine palmitoyltransferase 1; FOXO, forkhead box O; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase; LKB1, liver kinase B1; mTORC1, mechanistic target of rapamycin kinase complex 1; NAD+, nicotinamide adenine dinucleotide; NF-κB, nuclear factor kappa B subunit; NIK, NF-κB-inducing kinase; p300, E1A binding protein p300; PGC1α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha; Raptor, regulatory associated protein of mTOR complex 1; SIRT1, sirtuin 1; Smad3, SMAD family member 3; SREBP1/2, sterol regulatory element binding proteins 1 and 2; TAK1, transforming growth factor beta-activated kinase 1; TBK1, TANK binding kinase 1; TSC1/2, tuberous sclerosis proteins 1 and 2; ULK1, Unc-51-like autophagy activating kinase 1.
AMPK is believed to play a significant role in both ALD and NAFLD (53, 225, 238). AMPK regulates hepatic lipid homeostasis through multiple processes, including inhibition of lipogenesis and activation of fatty acid oxidation (FAO). Adenoviral overexpression of AMPKα1 in the liver reduces hepatic lipogenic gene expression and triglycerides in hyperlipidemic diabetic rats (181). Hepatic activation of AMPK also protects against diet-induced fatty liver in animal models (53). AMPK can inhibit lipogenesis by direct phosphorylation of acetyl-CoA carboxylases 1 and 2 (ACC1/2) (136). The ACC enzymatic product malonyl-CoA is not only a substrate for fatty acid synthase (FASN) but also an allosteric inhibitor for carnitine palmitoyltransferases (CPTs) that transport acyl-CoA into mitochondria for oxidation (56).
AMPK also directly phosphorylates and inhibits both sterol regulatory element binding proteins 1 and 2 (SREBP1 and SREBP2) to downregulate hepatic lipogenesis and cholesterol biosynthesis (108). In addition, AMPK phosphorylates both tuberous sclerosis complex subunit 2 (TSC2) and Raptor to inhibit mechanistic target of rapamycin kinase complex 1 (mTORC1), which activates hepatic lipogenesis by regulating SREBP maturation and activation (56, 198).
AMPK has an anti-inflammation function in the liver. Hepatic overexpression of constitutively active AMPK downregulates inflammatory genes in high-fat diet-treated mouse livers (53). AMPK also dampens hepatic inflammation through inhibition of the activity of nuclear factor kappa B subunit (NF-κB). On the one hand, AMPK can promote sirtuin 1 (SIRT1) activity by an increase of cellular NAD+ levels (17). As a result, SIRT1 suppresses the NF-κB transcriptional activity by deacetylation of its RelA/p65 subunit at Lys310 (222). On the other hand, AMPK can decrease the NF-κB signaling through the Unc-51 like autophagy activating kinase 1 (ULK1)–TANK binding kinase 1 (TBK1)–NF-κB-inducing kinase (NIK) axis in which AMPK phosphorylates ULK1 followed by TBK1 phosphorylation. Then TBK1 phosphorylates NIK to induce its degradation (45, 239).
AMPK can protect against hepatic apoptosis through direct phosphorylation and inhibition of procaspase 6 (238). Moreover, AMPK activation by small molecules such as A769662 reduces hepatic fibrosis in animal models (238). AMPK activation has been shown to suppress the expression of fibrogenic genes in hepatic stellate cells (HSCs). One of the potential mechanisms is that AMPK activation leads to dissociation of SMAD family member 3 (SMAD3) from its transcriptional coactivator p300 and proteasomal degradation of p300 (110). AMPK also inhibits HSC activation by control of the expression of vacuolar H+ adenosine triphosphatase and intracellular pH (126). Knockout of AMPKα1 in mice inhibits HSC proliferation but not fibrogenesis (33).
mTOR signaling
mTOR is a catalytic component of two protein complexes: mTORC1 and mTORC2. mTORC1 plays a critical role in metabolic homeostasis and cell growth, whereas mTORC2 activates a number of kinases, including AKT serine/threonine kinase (AKT), serum- and glucocorticoid-induced protein kinase, and protein kinase C (Fig. 2) (114).
FIG. 2.
mTOR signaling pathways in fatty liver disease. mTORC1 stimulates hepatic lipogenesis and inhibits lipid breakdown, but seems to play different roles in mild versus severe hepatic inflammation. mTORC2 also regulates lipogenesis, inflammation, and HSC activity. AKT1/2, AKT serine/threonine kinases 1 and 2; Arg, arginine; ATG13, autophagy-related gene 13; CASTOR1, cytosolic arginine sensor for mTORC1 subunit 1; EtOH, ethanol; EV, extracellular vesicle; GATOR1/2, GTPase-activating proteins 1/2 toward Rags complex; HIF1α, hypoxia inducible factor 1α; HSC, hepatic stellate cell; KC, Kupffer cell; Leu, leucine; LPS, lipopolysaccharide; Met, methionine; mTORC2, mechanistic target of rapamycin kinase complex 2; PI3K, phosphatidylinositol 3-kinase; RagA/B/C/D, Ras-related GTP binding proteins A/B/C/D; Rheb, Ras homologue enriched in brain; REDD1, protein regulated in development and DNA damage response 1; ROCK1, Rho-associated coiled-coil containing protein kinase 1; S6K1, ribosomal protein S6 kinase 1; SAM, S-adenosylmethionine; SAMTOR, S-adenosylmethionine sensor upstream of mTORC1; TFE3, transcription factor binding to IGHM enhancer 3; TFEB, transcription factor EB; UVRAG, UV radiation resistance associated.
mTORC1 promotes hepatic fatty acid and cholesterol biosynthesis by activation of SREBP1, SREBP2, and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) (44, 120, 221). Both ribosomal protein S6 kinase 1 (S6K1) and lipin-1 have been shown to be involved in the regulation of SREBPs (44, 221). mTORC2 also regulates hepatic lipogenesis via insulin-induced activation of AKT, glucokinase, and SREBP1c (60). SESN1/2/3 can activate AMPK and mTORC2 but inhibit mTORC1, and by doing so they promote hepatic lipid homeostasis (98, 193).
mTORC1 is a key negative regulator for autophagy (78). In the liver, autophagy helps maintain energy homeostasis and protect against cell injury or death by clearing damaged organelles and misfolded proteins. Autophagy also facilitates lipid droplet breakdown. mTORC1 inhibits the autophagy-initiating ULK complex by phosphorylation of autophagy-related gene 13 (ATG13) and ULK1/2 (78, 84). Autophagy is a double-edged sword in liver fibrosis. Most studies have demonstrated that increased autophagy activates HSCs by degrading lipid droplets (63). However, autophagy can also be used to reduce the production of fibrogenic extracellular vesicles (EVs) (52). mTORC1 overactivation in mesenchymal cells by deletion of TSC1 using Col1a2-Cre-ERT exacerbates CCl4-induced hepatic fibrosis but not under normal conditions (182). mTOR inhibitors sirolimus and everolimus reduce bile duct ligation-induced hepatic fibrosis in rats (156). The role of mTORC2 in HSCs remains unclear. One report suggests that mTORC2 might promote HSC proliferation and activation through AKTs (166).
The role of mTORC1 in hepatic inflammation remains debatable. Chronic mTORC1 activation can promote inflammation (24). Deletion of Raptor in macrophages improves insulin sensitivity and ameliorates adipose and hepatic inflammation in mice when challenged with a high-fat diet (75). However, under some other conditions such as NASH diets or autophagy deficiency, mTORC1 is required for the resolution of inflammation (145, 196).
Endoplasmic reticulum stress signaling
Endoplasmic reticulum (ER) is the major site of lipid synthesis in hepatocytes. Despite abundant ER, hepatocytes are very sensitive to the disruption of normal ER function by various ER stressors (95). ER stress is a major contributor to liver disease, including hepatic steatosis, inflammation, and fibrosis (Fig. 3) (95). In fatty liver disease, abnormal lipid accumulation often coexists with insulin resistance and proteostasis perturbation in hepatocytes. ER stress is often triggered by unfolded protein response (UPR) as misfolded or unfolded proteins are accumulated in the ER lumen (95). Worse yet, UPR can cause inflammasome activation and inflammation, and, in the case of nonresolvable ER stress, cell death (95). There are three major ER stress pathways that are mediated by the ER transmembrane proteins inositol-requiring enzyme 1 α (IRE1α), PRKR-like ER kinase (PERK), and activating transcription factor 6 α (ATF6α), respectively (95).
FIG. 3.
ER stress in fatty liver disease. There are three major ER stress signaling pathways mediated by PERK, IRE1α, and ATF6. ER stress can trigger inflammation, apoptosis, fibrogenesis, and other cellular events. ATF4/6, activating transcription factors 4 and 6; CHOP, CCAAT/enhancer-binding protein homologous protein; DNL, de novo lipogenesis; eIF2α, eukaryotic translation initiation factor 2 alpha; ER, endoplasmic reticulum; FAO, fatty acid oxidation; GADD34, growth arrest and DNA damage-inducible protein 34; HNRNPA1, heterogeneous nuclear ribonucleoprotein A1; IRE1α, inositol-requiring enzyme 1 α; JNK, JUN N-terminal kinase; miR, microRNA; NRF2, nuclear factor erythroid 2-related factor 2; PERK, PRKR-like endoplasmic reticulum kinase; PP1C, protein phosphatase 1C; PPARα, peroxisome proliferator-activated receptor α; SMAD2, SMAD family member 2; TXNIP, thioredoxin interacting protein; VLDL, very low-density lipoprotein; XBP1s, X-box binding protein 1 spliced; XBP1u, X-box binding protein 1 unspliced.
ER stress has been shown to cause hepatic steatosis (170). ER stress-induced hepatic steatosis can be regulated by multiple factors, and can happen through the regulation of lipogenesis, very low-density lipoprotein (VLDL) secretion, and FAO (170). The IRE1α–X-box binding protein 1 (XBP1) pathway plays an important role in the regulation of VLDL secretion and lipogenesis (206). Mice with hepatocyte-specific deletion of IRE1α develop hepatic steatosis and increase expression of several lipogenesis genes in the liver after the treatment with tunicamycin, an ER-stress inducer (235). In addition, XBP1 deletion decreases de novo hepatic lipogenesis, leading to reduced serum triglycerides, cholesterol, and free fatty acids through regulation of lipogenic genes, including ACC2 and stearoyl-CoA desaturase 1 (SCD1) (96). XBP1 deletion also triggers a feedback hyperactivation of IRE1α, thereby reducing plasma triglycerides and cholesterol in the XBP1 knockout mice (187). Activation of IRE1α increases the release of proinflammatory EVs from hepatocytes, whereas inhibition of IRE1α in the hepatocytes and in mice impairs hepatic inflammasome activation, liver injury, and hepatic cell apoptosis (95).
The PERK-eIF2α-ATF4 pathway also regulates hepatic steatosis. Hepatic overexpression of growth arrest and DNA damage-inducible protein 34 (GADD34), a regulatory subunit of protein phosphatase 1, which keeps eIF2α in a dephosphorylated state, protects mice from high-fat diet-induced obesity and hepatic steatosis (149). ATF4 deletion exhibits a protective effect against diet-induced hepatic steatosis in mice (104).
The ATF6α signaling pathway has also been shown to protect against hepatic steatosis. ATF6α knockout mice develop hepatic steatosis after the tunicamycin treatment probably due to the blockage of β-oxidation of fatty acids and the suppression of VLDL formation (25). ATF6α can activate peroxisome proliferator-activated receptor α (PPARα) to induce expression of FAO genes (25). ATF6α suppresses cholesterol biosynthesis by forming the ATF6-SREBP2-HDAC1 suppressive transcriptional complex (230). Furthermore, ATF6α knockout mice develop hepatic steatosis and glucose intolerance in association with increased expression of SREBP1c (197).
SESN2 can be induced by ER stress-activated CCAAT enhancer binding protein β (C/EBPβ) transcriptional factor during diet-induced obesity. Then SESN2 modulates UPR through suppression of mTORC1 and eventually suppresses the obesity-associated hepatic steatohepatitis (154). SESN2 can also be induced by ATF6 in response to ER stress, and SESN2 subsequently reduces UPR via inhibition of phosphorylation of JUN N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) and suppresses ER stress-induced hepatocyte apoptosis and liver injury (72).
ER stress has also been implicated in hepatic inflammation and fibrosis. IRE1α increases hepatic inflammation by two distinct mechanisms. The nuclease activity of IRE1α can increase the thioredoxin interacting protein (TXNIP) mRNA levels by reducing the TXNIP-targeting microRNA 17 (miR-17). The elevation of the TXNIP promotes the NLR family pyrin domain containing 3 (NLRP3)-mediated inflammasome activation (102). The kinase activity of IRE1α, in addition to phosphorylated eIF2α and ATF6, can activate the NF-κB-mediated inflammation (76). All three major ER stress pathways have been shown to increase hepatic fibrosis. PERK can directly phosphorylate and activate heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) to upregulate SMAD2 by downregulation of miR-18 that directly targets SMAD2 during the HSC activation (92). The IRE1α-XBP1 pathway also activates HSCs by triggering autophagy (64). The role of ATF6 in hepatic fibrosis is currently unclear.
Oxidative stress signaling
Generally, cells produce reactive oxygen species (ROS) and reactive nitrogen species at certain levels according to cellular conditions. As both oxidant species are closely related, we here use ROS to represent both. ROS production is involved in different enzymatic and nonenzymatic reactions. The ROS-producing enzymes include nicotinamide adenine dinucleotide (NAD) phosphate oxidase, xanthine dehydrogenase, cytochrome P450 2 subfamily E member 1 (CYP2E1), cyclooxygenases, and lipoxygenases (57).
In fatty liver disease, accumulation of saturated free fatty acids such as palmitates, diacylglycerol, ceramides, and cholesterol in the liver can induce lipotoxicity and oxidative stress, which can lead to hepatic inflammation, mitochondrial dysfunction, cell apoptosis, and liver fibrosis (Fig. 4) (57). Alcohol metabolism also generates ROS as by-product, especially by CYP2E1 (57). In the early stage of fatty liver disease, there is an increase in mitochondrial activity as a compensatory mechanism to protect hepatic cells against the harmful lipid overflow (125). As a result, this also leads to an increase in production and accumulation of ROS.
FIG. 4.
Oxidative stress in fatty liver disease. Multiple sources including free fatty acids, cholesterol, and ethanol can lead to the production of ROS, which may cause cell damage, inflammation, and even cell death. ADH, alcohol dehydrogenase; ALDH2, aldehyde dehydrogenase 2; AP-1, activator protein 1; ASK1, apoptosis signal regulating kinase 1; CAT, catalase; CYP2E1, cytochrome P450 family 2 subfamily E member 1; DAG, diacylglycerol; IKK, inhibitor of nuclear factor kappa B kinase; p38 MAPK, p38 mitogen-activated protein kinase; PA, palmitic acid; PM, plasma membrane; ROS, reactive oxygen species.
During the development of fatty liver disease, ER stress occurs and this also increases ROS production. One of the most significant causes in the ER stress-induced ROS production is intracellular calcium flux. Excess calcium flows into the cytoplasm, which can be absorbed by mitochondria, inducing the opening of the mitochondrial permeability transition pore and causing ROS production (46). When mitochondria become dysfunctional, excessive ROS are produced due to increased electron leakage, which in turns increases oxidative stress and damage to mitochondria. ROS can cause lipid peroxidation and mitochondrial membrane damage (125). Elevated biosynthesis through mitochondrial anaplerotic/cataplerotic pathways also contributes to oxidative stress and inflammation in the liver during NAFLD (173).
ROS regulate numerous signaling molecules that may lead to oxidative stress, cell damage, or cell death. Chronic plus binge alcohol consumption increases hepatic oxidative stress that triggers activation of apoptosis signal regulating kinase 1 (ASK1) and p38 MAPK signaling pathways and subsequently hepatic steatohepatitis (122). ROS also triggers hepatic inflammation by either direct activation of the NF-κB and JNK pathways or indirectly through the ER stress pathways (57). Oxidative stress also contributes to the development of hepatic fibrosis (240).
Inflammatory signaling
Inflammation can be a physiological or pathological response to tissue injury or infection by releasing proinflammatory cytokines, chemokines, and eicosanoids during cell defense and tissue repair (179). The triggers of hepatic inflammation can originate from either extrinsic or intrinsic factors (179). Adipose tissue can produce and release numerous hormones such as leptin, resistin, and visfatin, and proinflammatory cytokines such as tumor necrosis factor α (TNFα), interleukin 1β (IL1β), and interleukin 6 (IL6), and chemokines under pathological conditions such as obesity and diabetes (179). Obesity often leads to hyperlipidemia and hyperglycemia, and both can activate inflammatory pathways via toll-like receptors (TLRs), especially TLR4 (179).
Saturated fatty acid lauric acid can activate TLR4 by promotion of the receptor dimerization and recruitment to lipid rafts (213). TLR4 expression is increased in the liver of NASH patients and can be induced by palmitate (183). Upon activation, TLR4 stimulates the signaling cascade of interleukin 1 receptor-associated kinase (IRAK)–TNF receptor-associated factor 6 (TRAF6)–TAK1. From TAK1, there are at least three branches: p38 MAPK, JNK–activator protein 1 (AP-1), and inhibitor of NF-κB kinase (IKK)–NF-κB. IKK phosphorylates the NF-κB inhibitor and triggers its degradation. As a result, NF-κB is released from the protein complex and translocates to the nucleus for transcriptional activation of numerous inflammatory chemokine and cytokine genes in hepatocytes and hepatic immune cells (27).
Lipopolysaccharides (LPS) produced by intestinal microbiota also trigger hepatic inflammation mediated by TLR4, leading to proinflammatory cytokine synthesis by activation of AP-1 and NF-κB (19). Inflammatory cytokines such as TNFα, IL1β, and IL6 can also cause hepatic insulin resistance and inflammation through their respective receptors (19). IL6 activates Janus kinases (JAKs), which in turn activate signal transducer and activator of transcription 3 (STAT3) (175). TNFα acts on the TNF receptor (TNFR) to stimulate the signaling cascade of TNFR-associated death domain protein (TRADD)–TRAF2/5–IKK–NF-κB (121). IL1β binds to IL1 receptor (IL1R) to activate both p38 MAPK and JNK signaling pathways during the fatty liver disease development (Fig. 5) (90).
FIG. 5.
Inflammatory pathways in fatty liver disease. A number of factors, including LPS, PA, ATP, IL1β, IL6, and TNFα, can trigger inflammatory signaling cascades that lead to production of cytokines and chemokines and tissue injury. ASC, apoptosis-associated speck-like protein containing a CARD; ATP, adenosine triphosphate; Casp1, caspase-1; IL1β, interleukin 1 beta; IL1R, interleukin 1 receptor; IL6, interleukin 6; IL6R, interleukin 6 receptor; IRAK, interleukin 1 receptor associated kinase; JAK, Janus kinase; K+, potassium ion; MAP3K1, mitogen-activated protein kinase kinase kinase 1; MEK7, MAPK/ERK kinase 7; MKK4/6/7, mitogen-activated protein kinase kinases 4/6/7; MyD88, myeloid differentiation primary response protein 88; NLRP3, NLR family pyrin domain containing 3; P2X7, purinergic receptor P2X 7; STAT3, signal transducer and activator of transcription 3; TIRAP, TIR domain containing adaptor protein; TLR4, toll-like receptor 4; TNFα, tumor necrosis factor α; TNFR, tumor necrosis factor receptor; TRADD, tumor necrosis factor receptor type 1-associated death domain protein; TRAF2/5/6, tumor necrosis factor receptor-associated factors 2/5/6.
TGFβ signaling
In the liver, TGFβ is a key regulator of liver physiology and pathology, contributing to all stages of hepatic disease progression, from initial liver injury, inflammation, fibrosis, to cirrhosis and cancer (48). Generally, the TGFβ signaling transduction network can be divided into two categories: SMAD molecule-related signaling (canonical) and non-SMAD (noncanonical) pathways (48). TGFβ acts as dimers by first binding to a pair of type II TGFβ receptors (TGFβR2) and then recruiting two type I receptors (TGFβR1) to form a stable receptor complex. Next, TGFβR1 amplifies the TGFβ signal transduction by recruiting receptor-regulated SMAD proteins (SMAD2 and SMAD3) and phosphorylating them. Activated SMAD2 and SMAD3 can form a complex with SMAD4, then together translocate into the nucleus, bind to DNA, and regulate the expression of target genes, including fibrosis-related genes. In addition, TGFβ also induces expression of the inhibitory SMAD (SMAD7) to negatively regulate the signaling pathway (48).
Those non-SMAD pathways may initiate from either TGFβR1 or TGFβR2 depending on the proteins associated with them. When TGFβ stimulates its receptors, SHC adaptor protein A can be phosphorylated and then forms a protein complex with growth factor receptor bound protein 2 and SOS Ras/Rac guanine nucleotide exchange factor to activate the MAPK cascade (99). TRAF4/6 activated by TGFβR1 facilitate recruitment of TAK1 and subsequently activate JNK and p38 MAPK and NF-κB signaling (218). Phosphatidylinositol 3-kinase (PI3K)–AKT–mTOR can be activated by the TGFβ signaling in the regulation of hepatic cell survival and function. Small GTPases, including Ras homology family member A (RHOA), cell division cycle 42, and Ras-related C3 botulinum toxin substrate (RAC), can be activated by TGFβ in hepatic fibrogenesis (93). TGFβ can also activate the JAK–STAT3 signaling pathway in liver cells, including HSCs (190).
Sesn3 is an endogenous inhibitor of the TGFβ signaling pathway and it suppresses the TGFβ-induced hepatic fibrosis by inhibiting the TGFβ receptors through recruitment of inhibitory SMAD7 and sequestration of SMAD3 to the cytoplasm (Fig. 6) (67).
FIG. 6.
TGFβ signaling pathways in fatty liver disease. TGFβ can trigger canonical Smad2/3 signaling and multiple noncanonical pathways involving several kinases. ALK1/5, anaplastic lymphoma receptor tyrosine kinases 1 and 5; MAPK, mitogen-activated protein kinase; RAF, Raf proto-oncogene serine/threonine kinase; RAS, Ras proto-oncogene GTPase; RhoA, Ras homology family member A; Sesn3, sestrin 3; Smad2/3/4/7, SMAD family members 2/3/4/7; Smurf2, SMAD specific E3 ubiquitin protein ligase 2; TGFβ, transforming growth factor β.
Hippo signaling
The Hippo signaling pathway, especially downstream mediators Yes1-associated transcriptional regulator (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ/WWTR1), has been implicated in liver metabolism, inflammation, regeneration, fibrosis, and tumorigenesis (Fig. 7) (123). Upon activation, the mammalian homologues of Hippo—mammalian STE20-like protein kinases 1 and 2 (MST1/STK4 and MST2/STK3), phosphorylate and activate large tumor suppressor kinases 1 and 2 (LATS1/2), which subsequently phosphorylate cytoplasmic YAP. Phosphorylated YAP/TAZ can be bound by 14-3-3 and sequestered in the cytoplasm and subjected to proteasomal degradation. When the Hippo signaling is off, nonphosphorylated YAP/TAZ translocate to the nucleus and coactivate the TEA domain family of transcription factors (TEADs) to induce genes involved in hepatic cell proliferation, survival, and function (123). Vestigial-like 4 (VGLL4) antagonizes nuclear YAP by interfering with the YAP-TEAD interaction (123).
FIG. 7.
Hippo signaling pathway in fatty liver disease. Multiple extracellular and intracellular factors can regulate the Hippo signaling. YAP and TAZ are two major downstream transcriptional coactivators that facilitate the TEAD transcriptional activity. AMOT, angiomotin; ARID1A, AT-rich interaction domain 1A; GPCR, G protein-coupled receptor; KIBRA, kidney and brain protein; LATS1/2, large tumor suppressor kinases 1 and 2; MAP4K, mitogen-activated protein kinase kinase kinase kinase; MOB1A/B, MOB kinase activator 1 A/B; MST1/2, mammalian STE20-like protein kinases 1 and 2; NF2, neurofibromin 2; SAV1, Salvador family WW domain containing protein 1; TAZ, transcriptional coactivator with PDZ-binding motif; TEAD, TEA domain transcription factor; VGLL4, vestigial-like family member 4; YAP, yes1-associated transcriptional regulator.
A number of upstream regulators of the Hippo pathway have been identified, including low energy, low glucose, statin, lysophosphatidic acid (LPA), glucagon, epinephrine, bile acids (BAs), and mechanical forces (41). Low glucose and low energy induce the Hippo signaling and YAP/TAZ phosphorylation through AMPK (41). In contrast, high glucose activates YAP probably through O-glycosylation (41). YAP/TAZ can also be regulated by the mevalonate pathway through posttranslational modifications of the Hippo upstream regulators with geranylgeranyl (189). LPA acts through G protein-coupled receptors (GPCRs) to inhibit LATS1/2 and thus activate YAP/TAZ (226). Glucagon and epinephrine act on their respective GPCRs to activate LATS1/2 and thus inhibit YAP/TAZ (226). BAs can induce YAP activation by either downregulation of MST1/2 and LATS1/2 expression or an increase in MST1/2 phosphorylation (4). Matrix stiffness also regulates YAP activity. Increased extracellular matrix regulates F-actin polymerization in an RHOA-dependent manner to activate YAP/TAZ nuclear translocation (43).
The Hippo signaling pathway has been implicated in hepatic metabolism. YAP can inhibit hepatic gluconeogenesis by suppression of the PPARγ coactivator 1α (PGC1α) gene expression. YAP also upregulates expression of insulin receptor substrate 2 (73). Deletion of LATS2 or MST1 gene in mice leads to fatty liver disease by induction of SREBP2 and cholesterol biosynthesis (7). YAP activation also induces glutaminolysis (42). The Hippo pathway also plays a critical role in the pathogenesis of NASH, especially through regulation of hepatocyte stress and survival and HSC activation (42, 124, 133, 210). TAZ expression is specifically induced in hepatocytes of human NASH livers but not steatotic livers (210). Knockdown of hepatocyte TAZ by siRNA improves hepatic fibrosis in a diet-induced NASH mouse model (209).
Sirtuins, microRNAs, and DNA Methylation in Fatty Liver Disease
Sirtuins
Sirtuins are NAD+-dependent deacetylases or deacylases. There are seven sirtuins in mammals (SIRT1–7). SIRT1 and SIRT6 are two key nuclear sirtuins implicated in fatty liver disease (Fig. 8).
FIG. 8.
SIRT1 and SIRT6 in fatty liver disease. Among seven sirtuin family members, SIRT1 and SIRT6 are significantly implicated in the regulation of the pathogenesis of fatty liver disease. CAT, catalase; ChREBP, carbohydrate response element binding protein; FGF21, fibroblast growth factor 21; HMOX1, heme oxygenase 1; MT1/2, metallothioneins 1 and 2; MTF1, metal regulatory transcription factor 1; NCOA2, nuclear receptor coactivator 2; SOD, superoxide dismutase.
Sirtuin 1
SIRT1 has a salutary role in hepatic metabolic homeostasis, antioxidative stress, anti-inflammation, and antifibrosis. SIRT1 overexpression or activation by agonists protects against diet-induced NAFLD through an increase of fatty acid β-oxidation and a decrease of inflammation in the liver (50, 202). SIRT1 also protects against hepatic steatosis through induction of fibroblast growth factor 21 (FGF21) (107). SIRT1 catalytically inactive mutant mice exhibit higher SREBP1 and SCD1 levels and lower LKB1 and AMPK phosphorylation in the liver on a high-fat diet compared with wild-type (WT) mice (26). SIRT1 can directly inhibit SREBP1c through deacetylation (161). Repopulation of human SIRT1 knockdown-induced pluripotent stem cell-derived hepatocytes in rat livers induces hepatic steatosis and inflammation (28). Sirt1 liver-specific knockout mice develop high-fat diet-induced NAFLD due to dysregulation of PPARα and PGC1α (162). SIRT1 also suppresses inflammation by deacetylation of NF-κB (177). Hepatic Sirt1 deficiency in mice exacerbates alcohol-induced hepatic steatosis and inflammation through regulation of lipin-1 and SREBP1c (223).
Sirtuin 6
SIRT6 also plays a critical role in the regulation of hepatic homeostasis and function. Hepatic Sirt6 deletion leads to age-associated hepatic steatosis through activation of glycolysis and lipogenesis and inhibition of fatty acid β-oxidation partly due to an increase in histone H3 lysine 9 (K9) acetylation in the target gene chromatin regions (83). Hepatic Sirt6 deficiency also aggravates high-fat, high-fructose, diet-induced oxidation stress and fibrosis in the mouse liver via upregulation of BTB domain and CNC homologue 1 (Bach1) and downregulation of nuclear factor, erythroid 2 like 2 (Nrf2/Nfe2l2), a key antioxidative stress transcription factor. Moreover, SIRT6 promotes NRF2 binding to the target gene promoters in response to oxidative stress (77). SIRT6 also promotes hepatic fatty acid β-oxidation through activation of PPARα by recruitment and deacetylation of nuclear receptor coactivator 2 (NCOA2), a PPARα coactivator. In addition, SIRT6 also mediates the inhibitory effect of PPARα on SREBPs for cholesterol and triglyceride biosynthesis (142). SIRT6 can directly interact with SREBP1 and SREBP2 to suppress the expression of fatty acid and cholesterol biosynthetic genes (128, 192). SIRT6 suppresses ER stress-induced hepatic steatosis through deacetylation and subsequent degradation of the deacetylated XBP1s protein via the ubiquitin/proteasome system (11).
SIRT6 attenuates hepatic inflammation by either suppression of the NF-κB activity in the macrophage M1 polarization or inhibition of the transcriptional activity of c-Jun for the proinflammatory genes such as IL6 and C-C motif chemokine ligand 2 (CCL2/MCP1) (101, 215). SIRT6 also plays a critical role in the control of hepatic fibrosis during NAFLD. Hepatocyte- or HSC-specific deletion of Sirt6 in mice leads to diet-induced hepatic fibrosis through the TGFβ-SMAD2/3 signaling pathway, whereas overexpression of SIRT6 completely reverses NASH (233, 241). Hepatic Sirt6 deficiency also leads to ethanol-induced liver injury due to downregulation of metallothioneins 1 and 2 (Mt1 and Mt2) and elevation of oxidative stress in mouse livers. SIRT6 overexpression dramatically improves ALD. Mechanistically, SIRT6 induces hepatic Mt1 and Mt2 gene expression by deacetylation and activation of metal regulatory transcription factor 1 (MTF1) (82).
MicroRNAs
MicroRNAs are small noncoding RNAs, ∼20–24 nucleotides in length. MicroRNAs regulate gene expression primarily at the posttranscriptional levels. A number of microRNAs have been implicated in hepatic steatosis, including miR-21, miR-22, miR-27b, miR-29, miR-33a/b, miR-34, miR-122, miR-222, miR-223, miR-375, and miR-451. Numerous microRNAs are also involved in hepatic fibrosis, including miR-16, miR-21, miR-33, miR-34, miR-122, miR-125b, miR-155, miR-192, miR-222, and miR-223 (Fig. 9) (208).
FIG. 9.
microRNAs in fatty liver disease. Numerous microRNAs are involved in the regulation of hepatic steatosis, inflammation, and fibrosis. Here only a number of well-studied microRNAs are highlighted. ABCA1, ATP-binding cassette subfamily A member 1; ABCG1, ATP-binding cassette subfamily G member 1; AGPAT1, 1-acylglycerol-3-phosphate O-acyltransferase 1; BCL2, BCL2 apoptosis regulator; CCL2, C-C motif chemokine ligand 2; CCND1, cyclin D1; CD36, CD36 molecule; CDK6, cyclin-dependent kinase 6; CEBPB, CCAAT enhancer binding protein beta; CIDEC, cell death inducing DFFA-like effector C; COL3A1, collagen type III alpha 1 chain; CTGF, connective tissue growth factor; CXCL10, C-X-C motif chemokine ligand 10; DGAT1, diacylglycerol O-acyltransferase 1; FGF11, fibroblast growth factor 11; FGFR1, fibroblast growth factor receptor 1; FXR, farnesoid X receptor; G6PC, glucose-6-phosphatase catalytic subunit; G6PD, glucose-6-phosphate dehydrogenase; GLS, glutaminase; GNA12, guanine nucleotide-binding protein subunit alpha 12; GR, glucocorticoid receptor; HGF, hepatocyte growth factor; HMGA2, high-mobility group AT-hook 2; HMGCS1/2, 3-hydroxy-3-methylglutaryl-CoA synthases 1 and 2; IGF1, insulin-like growth factor 1; IGF1R, insulin-like growth factor 1 receptor; IL1RN, interleukin 1 receptor antagonist; KLF6, Kruppel-like factor 6; LAMP1, lysosomal-associated membrane protein 1; LXRA, liver X receptor alpha; MAP3K3, mitogen-activated protein kinase kinase kinase 3; MERTK, MER proto-oncogene tyrosine kinase; NDRG3, N-Myc downstream-regulated gene 3 protein; NFYC, nuclear transcription factor Y subunit gamma; PDGFC, platelet-derived growth factor C; PIM3, Pim-3 proto-oncogene serine/threonine kinase; PKM2, pyruvate kinase M2; PPARγ, peroxisome proliferator-activated receptor γ; PTEN, phosphatase and tensin homologue; RELB, RELB proto-oncogene; RICTOR, RPTOR independent companion of mTOR complex 2; SC4MOL, sterol-C4-methyl oxidase; SCD1, stearoyl-CoA desaturase 1; SMAD2/7, SMAD family members 2 and 7; SOCS1, suppressor of cytokine signaling 1; SP3, SP3 transcription factor; SR-BI, scavenger receptor class B member 1; VHL, von Hippel–Lindau tumor suppressor; WNT3A, Wnt family member 3A; ZEB2, zinc finger E-box binding homeobox 2.
miR-16
miR-16 has been shown to inhibit hepatic fibrosis through multiple targets, including guanine nucleotide-binding protein subunit alpha 12 (GNA12), SMAD2, and Wnt family member 3A (WNT3A) (86, 152).
miR-21
miR-21 has been extensively studied in hepatic metabolism and fibrosis. Numerous miR-21 target genes have been identified, including SMAD7, forkhead box O1 (FOXO1), HMGCR, and STAT3 (20, 214). However, some data suggest that miR-21 is not required for HSC activation and liver fibrogenesis (20).
miR-29
miR-29 has been shown to regulate hepatic lipogenesis and fibrosis. Regarding regulation of hepatic lipid metabolism, the miR-29 effects are rather complicated as both positive and negative factors are targets of miR-29, including SIRT1 and CD36 (68, 111). Overexpression of miR-29a improves diet-induced hepatic steatosis and fibrosis (111). miR-29a administration improves CCl4 and thioacetamide-induced hepatic fibrosis in mice. Collagen type I α1 (COL1A1) and platelet-derived growth factor C (PDGFC) are among the targets of miR-29a (129). Expression of miR-29 is suppressed by the TGFβ and LPS signaling pathways (168).
miR-33
miR-33 has been reported to regulate cholesterol homeostasis by targeting ATP-binding cassette transporter A1 (ABCA1), which is involved in high-density lipoprotein (HDL) biosynthesis and reverse cholesterol transport (143, 164). Short-term administration of anti-miR-33 oligonucleotides can increase HDL and decrease VLDL triglycerides in nonhuman primates. In addition, several FAO genes are also miR-33 targets (163). However, long-term silencing of miR-33 leads to elevated circulating and hepatic triglycerides in high-fat diet-treated mice due to increased lipogenesis. SREBP1 and nuclear transcription factor Y subunit gamma (NFYC) are among targets of miR-33 (55).
miR-122
miR-122 is highly abundant in the liver, which accounts for ∼70% of all hepatic microRNAs. miR-122 knockout mice develop hepatic steatohepatitis, fibrosis, and hepatocellular carcinoma (66, 195). miR-122 negatively regulates both lipogenic and FAO genes, including 1-acylglycerol-3-phosphate O-acyltransferase 1 (AGPAT1), SIRT1, and SIRT6 (47, 66, 119). Retinoic acid receptor-related orphan receptor A agonists can improve NASH by upregulating miR-122 in mice (22). miR-122 is decreased in the livers of ALD patients and mice. Overexpression of miR-122 in the liver protects against ALD partly through inhibition of hypoxia inducible factor 1 subunit alpha, a target of miR-122 (174).
miR-155
miR-155 is abundantly expressed in immune cells such as macrophages. miR-155 regulates hepatic lipid metabolism through suppression of lipogenic transcription factors such as C/EBPα and C/EBPβ, PPARγ, and liver X receptor α (LXRα) (203). miR-155 knockout mice exhibit decreased hepatic steatosis and fibrosis, but no improvement in hepatic inflammation after challenged with a methionine/choline-deficient diet (31). miR-155 can be induced by alcohol and LPS in macrophages and promotes inflammation (9, 10). In response to alcohol and LPS, miR-155 exacerbates hepatic inflammation through suppression of several negative regulators of the TLR4 signaling pathway, including IRAK3, SH2 domain-containing inositol 5′-phosphatase 1, suppressor of cytokine signaling 1 (SOCS1), and C/EBPβ in Kupffer cells (KCs) (9). miR-155 promotes alcohol-induced hepatic steatohepatitis and fibrosis through inhibition of PPARα, PPARγ, and C/EBPβ (10). miR-155 also targets lysosomal-associated membrane proteins 1 and 2 (LAMP1/2) to impair the autophagy and lysosome functions in ALD (8).
miR-192
miR-192 has been shown to mediate lipotoxicity-induced macrophage activation by targeting Rictor, a key component of mTORC2 (118). miR-192 inhibits lipogenesis in hepatocytes by targeting SCD1 and SREBP1 (112). miR-192 also aggravates oxidative stress-induced liver injury by targeting zinc finger E-box binding homeobox 2 (ZEB2) (169).
miR-223
miR-223 is highly enriched in neutrophils and it has been implicated in both ALD and NAFLD. Under the NAFLD conditions, hepatocytes can uptake miR-223-containing EVs from neutrophils via a VLDL-ApoE interaction to attenuate hepatic inflammation and fibrosis (62). In addition, macrophage-released miR-223 in the exosomes also reduces TAZ in hepatocytes to dampen hepatic fibrosis (61). Neutrophil miR-223 also attenuates hepatic inflammation and fibrosis by promoting conversion of macrophages from proinflammatory to restorative type (16). miR-223 regulates cholesterol homeostasis by targeting scavenger receptor class B member 1 and 3-hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1) (201). In a mouse model of chronic-plus-binge ethanol feeding, miR-223 is elevated in serum and neutrophils. Deletion of the miR-223 gene leads to increased liver injury, oxidative stress, and neutrophil infiltration. Mechanistically, miR-223 directly inhibits IL6 expression and subsequently downregulates the p47 neutrophil cytosolic factor 1 (p47phox/Ncf1) gene expression in neutrophils (105).
DNA methylation
DNA methylation, especially 5-methylcytosine in the genome, is modulated by DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B) and 2-oxoglutarate-dependent dioxygenases such as ten eleven translocation enzymes (TET1–3) (69). Some evidence suggests that DNA methylation is dynamically altered in the livers of ALD and NAFLD patients compared with normal controls (Fig. 10) (231). Hypermethylation of the PGC1α gene promoter has been shown to correlate with decreased PGC1α expression and mitochondrial function in the liver (188). Hypermethylation in several genes such as CPT1A and 24-dehydrocholesterol reductase (DHCR24) and hypomethylation in the dipeptidyl peptidase 4 (DPP4) gene have been associated with hepatic steatosis (13).
FIG. 10.
DNA methylation in fatty liver disease. In both human and animal NAFLD livers, both hypermethylation and hypomethylation sites in numerous genes have been associated with different disease conditions. CPT1A, carnitine palmitoyltransferase 1A; CYP27A1, cytochrome P450 family 27 subfamily A member 1; DHCR24, 24-dehydrocholesterol reductase; DNMT3A, DNA methyltransferase 3 alpha; DPP4, dipeptidyl peptidase 4; FASN, fatty acid synthase; FGFR2, fibroblast growth factor receptor 2; MAT1A, methionine adenosyltransferase 1A; NAFLD, nonalcoholic fatty liver disease; NPC1L1, NPC1-like intracellular cholesterol transporter 1; PRC1, protein regulator of cytokinesis 1; SLC9A3R1, SLC9A3 regulator 1; SLC27A5, solute carrier family 27 member 5; SLC43A1, solute carrier family 43 member A1; SLC51A, solute carrier family 51 subunit alpha; TG, triglyceride; TRIM4, tripartite motif containing 4; TUBA1B, tubulin alpha 1b.
In the NASH livers, DNA methylation in the ATP citrate lyase (ACLY) gene CpG sites is decreased, whereas it is increased in the insulin-like growth factor 1 (IGF1) and IGF binding protein 2 (IGFBP2) genes (2). Hypermethylation in the genes of cytochrome P450 family 27 subfamily A member 1 (CYP27A1), NPC1 like intracellular cholesterol transporter 1 (NPC1L1), and solute carrier family 51 subunit α (SLC51A) has been associated with NASH (139). Hypomethylation in the genes of protein regulator of cytokinesis 1 (PRC1) and tripartite motif containing 4 (TRIM4) has been associated with NASH (194). When severe and mild NAFLD liver samples are compared, several CpG sites in the promoters of PPARα and PPARδ genes are hypermethylated, whereas the TGFB1 and PDGFA gene promoters are hypomethylated (231).
When liver samples of normal control and ALD patients are analyzed for DNA methylation profiles, both PPARα and PPARδ gene promoters are hypermethylated in the ALD livers, whereas the COL1A1 gene promoter is hypomethylated (231). Hepatic TET2 protein levels are decreased and DNMT3B protein levels are increased in the ALD patients compared with normal controls (151).
In rat HSCs, DNMT3A and DNMT3B are induced during transdifferentiation to myofibroblasts (151). Hypermethylation in the genes of methionine adenosyltransferase 1A (MAT1A), PPARα, and PPARγ has been associated with hepatic fibrosis (137). Hypomethylation of the genes of caspase-1 (CASP1) and fibroblast growth factor receptor 2 (FGFR2) has been associated with hepatic fibrosis (137). Interestingly, a dual G9a/DNMT1 inhibitor—CM272, suppresses the TGFβ-induced fibrogenesis in human LX-2 HSCs and improves hepatic fibrosis in CCl4 and bile duct ligation mouse models (12). Although its protein levels are not altered in human fibrotic livers, the chromatin occupancies of DNMT3A at the SREBP1 and FASN gene promoters are decreased in the livers of human steatosis and NASH patients (87).
Transcriptional Regulation of Hepatic Steatosis, Inflammation, and Fibrosis
Carbohydrate response element binding protein
Carbohydrate response element binding protein (ChREBP) is a basic/helix–loop–helix/leucine zipper transcription factor that is responsive to carbohydrate load, especially glucose. There are two isoforms – ChREBPα and ChREBPβ. ChREBPα has a low-glucose inhibitory domain (LID) and a glucose-response activation conserved element in the N-terminal of the protein (106); however, ChREBPβ does not have the LID motif (106). Glucose metabolites facilitate the nuclear translocation and transcriptional action of ChREBP through binding to a carbohydrate response element (ChoRE) in collaboration with Max-like protein X. Numerous ChREBP target genes have at least one ChoRE in their promoter region, for example, pyruvate kinase L/R (PKLR), FASN, ACC1, and SCD1 (148). ChREBP can be activated by O-GlcNAcylation and lysine acetylation (220).
Deletion of the ChREBP gene in mice leads to reduced glycolysis and lipogenesis (70). In an ob/ob mouse model, liver-specific knockdown of ChREBP using adenovirus-mediated shRNAs improves hepatic steatosis and insulin sensitivity due to reduced hepatic lipogenesis (37). Hepatic ChREBP overexpression in mice leads to increased hepatic steatosis but improved insulin signaling on a high-fat diet (14). ChREBP also modulates an adaptive response to high-fructose diet by induction of FGF21 to control fructose metabolism and lipogenesis in the liver (51). In response to a high-fructose diet, ChREBP can protect liver from overproduction of cholesterol and CCAAT/enhancer-binding protein homologous protein (CHOP)-mediated hepatic injury through an interaction with SREBP2 and promotion of its degradation (Fig. 11) (232).
FIG. 11.
Key transcription factors in the regulation of hepatic lipogenesis. A number of key transcription factors that have been implicated in the regulation of hepatic lipogenesis are highlighted here: ChREBP, LXR, SREBP1c, and FOXO1. AAs, amino acids; Ac, acetyl; ATG14, autophagy-related gene 14; ATGL, adipose triglyceride lipase; BAs, bile acids; CDKN1B, cyclin-dependent kinase inhibitor 1B; CFLAR, CASP8 and FADD apoptosis regulator; CYP7A1, cytochrome P450 family 7 subfamily A member 1; ERK, extracellular signal-regulated kinase; GCK, glucokinase; GlcNAc, N-acetyl-d-glucosamine; HSL, hormone-sensitive lipase; IKBE, NFKB inhibitor epsilon; LPL, lipoprotein lipase; OGT, O-linked N-acetylglucosamine transferase; PKA, protein kinase A; PP2A, protein phosphatase 2A; RAs, retinoic acids; RXR, retinoid X receptor.
Liver X receptor
The LXR family has two members—LXRα and LXRβ. Whereas LXRβ is ubiquitously expressed, LXRα is abundant in metabolically active tissues, including adipose tissue and liver (204). LXRs function as heterodimers with retinoid X receptor α (RXRα). Transcriptional activity of LXRs is induced by elevated levels of cholesterol or other endogenous ligands in the cell. LXRs regulate multiple lipid metabolic processes, including lipogenesis, cholesterol absorption, transport, and conversion to BAs (204). Regarding lipogenesis, LXRs activate the transcription of the SREBP1, ChREBP, FASN, and SCD1 genes (21, 165, 204).
LXRs play an important role in cholesterol metabolism. Cytochrome P450 family 7 subfamily A member 1 (CYP7A1), the rate-limiting enzyme in the biosynthesis of BAs, is a direct transcriptional target of LXRs (158). LXRs also induce expression of biliary cholesterol excretion genes such as ATP-binding cassette subfamily G members 5 and 8 (ABCG5/8) (228). Two reverse cholesterol transport genes ABCA1 and ABCG1 are also regulated by LXRs (80, 200). LXRs have been implicated in the NAFLD pathogenesis. LXR exerts anti-inflammation function through regulation of the activity of KCs and infiltrated macrophages and cholesterol homeostasis in the liver (158). However, data on targeting LXR for NAFLD are not very consistent as both salutary and harmful effects have been reported (Fig. 11) (88).
Sterol regulatory element binding proteins
SREBPs are synthesized as membrane proteins that are situated in the ER. In the absence of sterols, SREBP interacts with the SREBP-cleavage activating protein (SCAP) and then translocates to the Golgi complex where the SREBP precursor is processed by proteases S1P and S2P to generate a mature form of SREBP transcription factor. There are three members of SREBP—SREBP1a, SREBP1c, and SREBP2. Proteolysis of SREBP1 is not strongly regulated by sterols, but the maturation process is inhibited by polyunsaturated fatty acids and promoted by insulin and high levels of glucose (185).
SREBP1c activates lipogenic genes such as ACC1, FASN, and SCD1 (184). Deletion of the SREBP1 gene in ob/ob mice partially improves hepatic steatosis, whereas deletion of the SCAP gene in ob/ob mice or high-fat-induced obese mice prevents hepatic steatosis (132, 217). SREBP1c levels are elevated in the liver of human hepatic steatosis patients (141). SREBP2 mRNA and free cholesterol levels are increased in the livers of human NASH patients compared with normal controls (131). The SREBP2 upregulation may be attributed to hyperinsulinemia, inflammatory cytokines, or miR-122 downregulation (Fig. 11) (138).
Forkhead box O
FOXO transcription factors are members of the O subgroup of the Forkhead family that contain a conserved DNA binding domain. There are four FOXO genes—FOXO1/3/4/6 in mammals. FOXO transcriptional activities are tightly regulated by posttranslational modifications that control either cytosolic or nuclear localization of FOXOs. Phosphorylation at several conserved sites (FOXO1-T24/S256/S319, FOXO3-T32/S253/S315, FOXO4-T32/S197/S262), primarily catalyzed by Akt, induces the nuclear exclusion of FOXOs (39).
FOXOs regulate glucose homeostasis through promotion of hepatic gluconeogenesis and inhibition of glycolysis (40, 59). FOXOs also regulate hepatic lipid metabolism by activating lipolysis and lipophagy through upregulation of adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), lipoprotein lipase (LPL), CPT1A, ATG5, and ATG14 (39, 216, 236). FOXO1 also suppresses de novo lipogenesis by downregulation of SREBP1 and other lipogenic genes (36, 237). Deletion of hepatic FOXO1/3 or FOXO1/3/4 in mice leads to hepatic steatosis (191, 234). FOXO1/3/4 liver-specific knockout mice develop severe NASH on a high-fat, high-cholesterol diet (Fig. 11) (153).
FOXO1 plays a significant role in the regulation of macrophage activity. FOXO1 promotes apoptosis of insulin-resistant macrophages by inhibition of p65 NF-κB via induction of NF-κB inhibitor ɛ (IKBE) (180). Under obesity and insulin resistance conditions, FOXO1 and TLR4 form a feedback circuitry to modulate an inflammatory response in which FOXO1 transcriptionally activates TLR4, and subsequently, the TLR4 pathway inhibits the FOXO1 transcriptional activity in macrophages (Fig. 11) (49).
FOXO1 can inhibit the proliferation and transdifferentiation of HSCs through induction of p27Kip1 encoded by cyclin-dependent kinase inhibitor 1B (CDKN1B) and manganese-containing superoxide dismutase encoded by superoxide dismutase 2 (SOD2). FoxO1+/− mice are more susceptible to hepatic fibrosis induced by bile-duct ligation compared with WT mice (1). FOXOs are negatively regulated by the PI3K-AKT signaling pathway. In human HSC line LX-2 cells, TNF-related apoptosis inducing ligand inhibits the PI3K/AKT signaling and thus activates FOXOs. As a result, expression of an apoptosis suppressor CASP8 and FADD like apoptosis regulator (CFLAR) is downregulated and this leads to HSC apoptosis through several apoptosis-activating molecules such as caspase-3, caspase-8, and BH3 interacting domain death agonist (155). Liver-specific deletion of FOXO1/3/4 in mice leads to severe hepatic steatosis and fibrosis with elevated expression of Col1a1, Pdgfrb, Tgfb1, and TIMP metallopeptidase inhibitor 1 (Timp1) in the liver on a high-fat diet compared with that in WT controls (Fig. 11) (153).
Peroxisome proliferator-activated receptor α
Among three PPAR nuclear receptors, expression of PPARα is the most abundant in the liver. It is responsible for the clearance of lipids in the liver through regulation of numerous genes involved in FAO, including CPT1A and acyl-CoA oxidase 1 (ACOX1) (130). PPARα also controls plasma triglyceride levels through upregulation of LPL (176). Moreover, PPARα affects lipogenesis through regulation of expression of malonyl-CoA decarboxylase, an enzyme that degrades fatty acid synthesis precursor malonyl-CoA (97). PPARα whole-body or liver-specific knockout mice exhibit severe steatohepatitis after either fasting or feeding with a high-fat diet (Fig. 12) (81). PPARα has anti-inflammation function in the liver. One of the main mechanisms is to transrepress inflammatory transcription factors such as NF-κB, AP-1, and STATs (157). PPARα also controls the duration of inflammation by regulating the breakdown of its ligand leukotriene B4, an inflammatory inducer (38). PPARα-null macrophages have increased NF-κB, JNK, and p38 MAPK activities and inflammatory mediators compared with their WT counterparts (Fig. 12) (30).
FIG. 12.
PPARs and FXR in fatty liver disease. Main functions of PPARα, PPARδ, PPARγ, and FXR in the liver are summarized here. 9-HODE, 9-hydroxyoctadecadienoic acid; 15-HETE, 15-hydroxyeicosatetraenoic acid; ACOX1, acyl-CoA oxidase 1; ACTA2, actin alpha 2, smooth muscle; APOA4, apolipoprotein A4; CD68, CD68 molecule; CEBPD, CCAAT enhancer binding protein delta; CLEC4F, C-type lectin domain family 4 member F; COL1A1, collagen type I alpha 1 chain; CYP8B1, cytochrome P450 family 8 subfamily B member 1; ETS1, ETS proto-oncogene 1; FABP1, fatty acid binding protein 1; FAs, fatty acids; FBP1, fructose-bisphosphatase 1; HDAC3, histone deacetylase 3; IKBKE, inhibitor of nuclear factor kappa B kinase subunit epsilon; LIPG, lipase G; LTB4, leukotriene B4; M2 φ, M2 macrophage; NCOR, nuclear receptor corepressor; PKLR, pyruvate kinase L/R; PPARδ, peroxisome proliferator-activated receptor δ; SHP, small heterodimer partner; SUMO, sumoylation; VLDLR, very low-density lipoprotein receptor.
Peroxisome proliferator-activated receptor δ
PPARδ, structurally very similar to PPARα, is ubiquitously expressed in most tissues. Like the other members of the PPAR family, it forms a heterodimer with RXR and regulates expression of its target genes by binding to specific PPAR response elements. It is expressed in all cell types in the liver (229). In contrast to PPARα-null mice, PPARδ knockout mice have no significant alterations in plasma free fatty acids and β-hydroxybutyrate and hepatic triglycerides and glycogen. Transcriptional profiling reveals that PPARδ positively regulates PKLR and fructose-bisphosphatase 1 (FBP1) genes in glucose metabolism and apolipoprotein A4 (APOA4), LIPG, and VLDLR for lipoprotein metabolism, and negatively regulates inflammatory genes such as STAT1, IKBKE, and CD68 (Fig. 12) (172). Hepatic overexpression of PPARδ in mice decreases fasting blood glucose levels but increases hepatic glycogen and triglyceride deposition. However, this does not lead to hepatic injury possibly due to an increase in monounsaturated fatty acids and a decrease in saturated fatty acids (116). PPARδ exerts protective effects against hepatic inflammation. PPARδ promotes alternative M2 activation of hepatic KCs and ameliorates obesity-induced inflammation (147). Activation of PPARδ by specific agonists reduces expression of inflammatory and ER stress genes in the liver (Fig. 12) (100).
Peroxisome proliferator-activated receptor γ
PPARγ expression is low in the normal liver, but it is elevated in the liver of NAFLD patients (159). PPARγ promotes steatosis probably due to its role in lipogenesis. PPARγ knockout mice are protected from hepatic steatosis but develop severe hyperglycemia and insulin resistance (Fig. 12) (54, 134).
PPARγ has anti-inflammatory function, especially through control of macrophage activation. In macrophages, activation of PPARγ downregulates inflammatory genes partly by antagonizing transcription factors such as AP-1, NF-κB, and STAT (167). PPARγ also induces monocyte differentiation toward anti-inflammatory M2 macrophages. PPARγ activation has been shown to protect from hepatic inflammation through inhibition of M1 macrophage polarization and promotion of alternative M2 activation under high-fat diet conditions (Fig. 12) (146).
PPARγ plays a critical role in the maintenance of HSC quiescence. Deletion of PPARγ in HSCs using an aP2-Cre leads to exacerbated hepatic fibrosis in mice when challenged with CCl4 (135). PPARγ is downregulated during the HSC activation and re-expressed during the fibrosis resolution. Knockout of PPARγ specifically in mouse HSCs leads to downregulation of C/EBPδ, SREBP1, and ETS proto-oncogene 1 (ETS1). After the CCl4 treatment, HSC-specific PPARγ-deficient mice have 40% more hepatic fibrosis and are 30% slower in resolution of hepatic fibrosis than that in WT mice. Administration of rosiglitazone, a PPARγ agonist, improves the hepatic fibrosis regression in WT mice but not in PPARγ HSC-specific knockout mice (Fig. 12) (117).
Farnesoid X receptor
Farnesoid X receptor (FXR) is a receptor for BAs, with chenodeoxycholic acid being the most potent followed by deoxycholic acid, cholic acid, and lithocholic acid. FXR plays a major role in the feedback regulation of BA biosynthesis by suppression of the CYP7A1 and cytochrome P450 family 8 subfamily B member 1 (CYP8B1) genes via small heterodimer partner and FGF19 (91, 186). In addition, activation of FXR also regulates glucose and lipid homeostasis (85). FXR knockout mice have elevated levels of hepatic triglycerides and serum triglycerides and cholesterol (186). FXR inhibits hepatic lipogenesis by suppression of the transcriptional activity of LXR and consequently the expression of SREBP1c and ChREBP (18, 211). FXR activation also induces the PPARα gene expression for promotion of FAO (Fig. 12) (160).
Activator protein-1
AP-1 is a transcription factor that is composed of a homodimer or a heterodimer with at least one protein from the Jun family (c-Jun, JunB, and JunD) and another from the Fos family (c-Fos, FosB, Fra1, Fra2, and ΔFosB) or ATFs (65). Expression of c-Jun is increased in both hepatocytes and nonparenchymal liver cells (NPLCs) in the livers of NASH patients compared with hepatic steatotic patients (178). Conditional knockout of c-Jun in mouse livers using Alfp-Cre or Mx1-Cre reveals that c-Jun in the hepatocytes promotes cell survival and restrains biliary ductal reaction, whereas c-Jun in NPLCs promotes ductal reaction and hepatic fibrogenesis via regulation of osteopontin (OPN) and CD44 (Fig. 13) (178).
FIG. 13.
Key transcription factors in the regulation of hepatic inflammation. Numerous transcription factors are involved in the regulation of hepatic inflammation. AP-1, NF-κB, PU.1, STAT1, and KLF4 are highlighted here. ARG1, arginase 1; CCL5, C-C motif chemokine ligand 5; CD44, CD44 molecule; C/EBPβ, CCAAT enhancer binding protein β; FIZZ1, found in inflammatory zone 1; IFNγ, interferon gamma; KLF4, Kruppel-like factor 4; MCPIP, monocyte chemotactic protein-induced protein; MRC1, mannose receptor C-type 1; NASH, nonalcoholic steatohepatitis; NPLCs, nonparenchymal liver cells; OPN, osteopontin; PU.1, transcription factor PU.1.
Nuclear factor kappa B subunit
NF-κB is composed of a heterodimer or homodimer of the Rel family members encoded by the NFKB1, NFKB2, REL, RELA, and RELB genes. It is often activated in various chronic liver diseases, including ALD and NAFLD, and a number of proinflammatory cytokine and chemokine genes such as IL1β, IL6, TNFα, CCL2, and CCL5 are often upregulated by NF-κB (121). NF-κB plays a significant role in the development of methionine/choline-deficient (MCD) diet-induced NASH in mice (34). Pharmacological inhibition of IKK2, an upstream activator of NF-κB, in a high-sucrose diet-induced NASH model, remarkably reduces hepatic steatosis and inflammation (Fig. 13) (15). The proinflammatory activity of NF-κB in macrophages can be suppressed by SIRT1 via deacetylation (177).
Transcription factor PU.1
PU.1 transcription factor, a member of the ETS family, plays an important role in the differentiation of various immune cells, including macrophages, neutrophils, dendritic cells, T lymphoid cells, and B lymphoid cells (103). Knockdown or inhibition of PU.1 significantly suppresses hepatic inflammatory genes, including IL1β, IL6, and TNFα, especially in macrophages, in a high-fat diet-induced NAFLD or db/db mouse model (Fig. 13) (115).
Signal transducer and activator of transcription 1
STAT1 plays a different role than STAT3 in diet-induced NASH. Hepatic Stat1 and Stat3 are both activated in the liver of C57BL/6 mice after feeding with high-fat diets. Reduction of Stat1, but not Stat3, levels in the liver impedes T cell infiltration and NASH development in mice, and several inflammatory and fibrotic genes such as smooth muscle actin α2 (Acta2), Tgfb1, Il1b, Il6, and Tnf are downregulated (Fig. 13) (58).
Kruppel-like factor 4
Kruppel-like factor 4 (KLF4) is a zinc finger-containing transcription factor and it regulates macrophage polarization (109). After exposure to IL4, KLF4 is induced as well as STAT6. Through upregulation of MCP1-induced protein (MCPIP), KLF4 promotes M2 polarization via PPARγ and C/EBPβ and suppresses M1 polarization via NF-κB (79). In an ALD mouse model, knockdown of KLF4 by siRNA increases M1 macrophage markers, while KLF4 overexpression decreases M1 macrophage markers and increases M2 macrophage markers such as arginase 1 (Arg1) and Il10 (171). Sumoylation of KLF4 induced by IL4 promotes macrophage M2 polarization (Fig. 13) (205).
ETS proto-oncogene 1
Both in vitro and in vivo data suggest that ETS1 plays a critical role in the maintenance of HSC quiescence (117). ETS1 gene expression is high in fresh quiescent rat HSCs but diminishes after 7 days of culture (activation) (89). Knockdown of Ets1 in mouse primary HSCs by shRNA leads to upregulation of proliferation genes such as cyclin-dependent kinases 2 and 4 (Cdk2/4) and cyclin D1 (Ccnd1), fibrosis genes such as Col1a1, Timp1, and lysyl oxidase like 2 (Loxl2), and inflammation genes such as Il1b, Il6, C-X-C motif chemokine ligand 1 (Cxcl1), and Cxcl5, and downregulation of quiescence-associated genes such as Pparγ and insulin-induced gene 1 (Insig1). Knockout of one allele of the Ets1 gene in mouse HSCs using Lrat-Cre leads to worse hepatic fibrosis than WT mice after the CCl4 treatment (117). In addition to the control of the HSC quiescent state, ETS1 also functions against fibrosis through other mechanisms. For example, ETS1 increases matrix metallopeptidase 1 (MMP1) gene expression in response to hepatocyte growth factor (150). In fibroblasts, ETS1 plays an antagonistic role against the TGFβ-induced fibrotic activity potentially by sequestering SMAD transcription factors from their target genes (Fig. 14) (32).
FIG. 14.
Key transcription factors in the regulation of hepatic fibrosis. Numerous transcription factors have been implicated in the regulation of hepatic fibrosis. ETS1, GATA4, TCF21, β-catenin, SMAD2/3, YAP, and TAZ are highlighted here. BIRC5, baculoviral IAP repeat containing 5; CXCL5, C-X-C motif chemokine ligand 5; CYR61, cysteine-rich angiogenic inducer 61; FN1, fibronectin 1; FZD, frizzled class receptor; GATA4/6, GATA binding proteins 4/6; GLI2/3, GLI family zinc finger 2/3; IHH, Indian hedgehog signaling molecule; INSIG1, insulin-induced gene 1; LOXL2, lysyl oxidase-like 2; LRP5/6, LDL receptor-related proteins 5/6; MMP7/14, matrix metallopeptidases 7/14; PAI-1, plasminogen activator inhibitor 1; PDGFB, platelet-derived growth factor B; PDGFRB, platelet-derived growth factor receptor B; SMAD2/3, SMAD family members 2/3; TCF21, transcription factor 21; TGFBR1, transforming growth factor beta receptor 1; TIMP1, TIMP metallopeptidase inhibitor 1.
GATA binding protein 4
GATA binding protein 4 (GATA4) plays a protective role in hepatic fibrogenesis (35). In conditional heterozygous Gata4 flox/+:G2-Cre mice, CCl4 treatment leads to severe fibrogenesis compared with CCl4-treated WT mice. Transfection of GATA4 in LX-2 cells downregulates expression of fibrosis marker genes such as ACTA2 and collagen (35). Knockdown of Gata4 in mouse HSCs decreases Pparγ and increases Col1a1, Timp1, Loxl2, Il6, Ccl5, and Cxcl5 (117). Interestingly, endothelial Gata4 also regulates hepatic fibrosis through an angiocrine signaling manner, specifically by suppressing the Pdgfb gene expression in liver sinusoidal endothelial cells (Fig. 14) (212).
Transcription factor 21
Transcription factor 21 (TCF21) is a basic helix-loop-helix transcription factor and plays a role in the regulation of cell fate and differentiation during development (5). Through a cell-based transcription factor screening, TCF21 has been identified as a suppressor of the HSC activation. Tcf21 expression is downregulated during fibrogenesis and re-expressed during fibrosis regression in mice treated with CCl4. AAV6-mediated TCF21 overexpression protects against CCl4-induced hepatic injury and fibrosis. Mechanistically, TCF21 suppresses the TGFβ-induced fibrogenic gene program, including ACTA2 and COL1A1 (Fig. 14) (144).
β-Catenin
β-Catenin has been implicated in HSC activation and hepatic fibrosis. Gene expression profiling suggests that Wnt signaling genes such as frizzled class receptor 2 (FZD2), WNT4, and WNT5 are upregulated in the activated rat HSCs compared with control quiescent cells (74). WNT3A promotes HSC activation and survival, whereas secreted frizzled-related protein 1 (sFRP1) inhibits both (140). sFRP5 also inhibits HSC proliferation and migration in vitro and attenuates CCl4-induced hepatic fibrosis in mice (23). LDL receptor-related protein 6 (LRP6), a coreceptor for WNT, is elevated in livers of fibrotic patients. LRP6 gene knockout in cultured rat HSCs and in CCl4-treated rats reduces HSC activation and hepatic fibrosis (Fig. 14) (227).
SMAD3
SMAD3 is a key transcription factor mediating the TGFβ profibrogenic activity. Numerous fibrotic genes, including COL1A1 and TIMP1, are direct targets of SMAD3 (199). In vitro data suggest that TGFβ activates SMAD2 in early cultured HSCs and SMAD3 in activated myofibroblasts (113). Ablation of the Smad3 gene in mice leads to resistance to dimethylnitrosamine-induced liver fibrosis (94). After acute liver injury, both SMAD2 and SMAD3 induce the collagen gene expression stimulated by TGFβ and PDGF. During chronic liver disease, SMAD2 and SMAD3 may be regulated differently in response to TGFβ and PDGF (224). SMAD3 transcriptional activity is modulated by lysine acetylation primarily by p300 and deacetylation by SIRT1 and SIRT6 (71, 241). HSC-specific knockout of Sirt6 leads to hyperacetylated Smad2/3 and exacerbated hepatic fibrosis (233, 241). Aryl hydrocarbon receptor also suppresses HSC activation and hepatic fibrosis through disruption of the interaction between SMAD3 and β-catenin (Fig. 14) (219).
YAP and TAZ
As key downstream mediators of the Hippo signaling pathway, both YAP and TAZ/WWTR1 have been implicated in hepatic fibrosis. YAP is activated in HSCs in CCl4-treated mice and fibrotic human patients (124). Expression of YAP target genes such as connective tissue growth factor (CTGF) and ankyrin repeat domain 1 (ANKRD1) is increased during the initial stage of HSC activation in vitro. Knockdown of YAP or pharmacological inhibition of YAP blocks HSC activation in vitro. Administration of verteporfin, a chemical compound that disrupts the YAP/TEAD complex, to CCl4-treated mice remarkably improves hepatic fibrosis (124). Overexpression of YAP in hepatocytes induces hepatic inflammation and fibrosis as early as 2 weeks. Deletion of Yap alone or together with Taz from hepatocytes improves CCl4-induced inflammation and fibrosis. Cysteine-rich angiogenic inducer 61 (CYR61/CCN1), a transcriptional target of YAP and TAZ, is shown to mediate, at least partially, the YAP/TAZ effects on hepatic inflammation and fibrosis (133). YAP has been shown to mediate the integrin b1-triggered fibrogenesis (127). YAP activation also drives HSC transdifferentiation by induction of glutaminolysis (Fig. 14) (42).
TAZ is highly expressed in NASH livers (210). TAZ undergoes similar regulation to YAP by LATS1/2 with regard to its phosphorylation and cellular localization. Unlike YAP, the role of TAZ in HSCs remains elusive. Increasing evidence suggests that hepatocyte TAZ can indirectly promote HSC activation by induction and secretion of Indian hedgehog, an inducer of HSC activation. TAZ expression is elevated in hepatocytes in human and mouse livers during the transition from steatosis to NASH (210). Further study suggests that cholesterol is a trigger for the upregulation of TAZ in the NASH hepatocytes through a soluble adenylyl cyclase—protein kinase A—Ca2+—RHOA pathway that prevents the proteasomal degradation of TAZ (207). Administration of siRNA against hepatocyte Taz attenuates hepatic injury, inflammation, and fibrosis in a diet-induced NASH mouse model and also partially reverses the established NASH (209). Ceramide has also been shown to inhibit the YAP/TAZ signaling and hepatic fibrosis by promoting phosphorylation and proteasomal degradation of YAP/TAZ (Fig. 14) (3).
Concluding Remarks
As fatty liver disease is a complex metabolic disease, a variety of factors ranging from environmental (diet, alcohol, life style, and so forth), genetic (common and rare gene mutations), and epigenetic (DNA methylation, histone modifications, noncoding RNAs, and others) should be considered for both research and clinical purposes. New technologies such as next-generation sequencing, proteomics, and metabolomics are expected to further enhance our understanding of the pathogenesis and potential therapeutic targets for the fatty liver disease. It is foreseeable that effective therapeutics for this very common liver disease will become available in the near future.
Abbreviations Used
- 9-HODE
9-hydroxyoctadecadienoic acid
- 15-HETE
15-hydroxyeicosatetraenoic acid
- AA
amino acid
- ABCA1
ATP-binding cassette subfamily A member 1
- ABCG1/5/8
ATP-binding cassette subfamily G members 1, 5, and 8
- Ac
acetyl
- ACC1/2
acetyl-CoA carboxylases 1 and 2
- ACOX1
acyl-CoA oxidase 1
- ACTA2
actin alpha 2, smooth muscle
- ADH
alcohol dehydrogenase
- AGPAT1
1-acylglycerol-3-phosphate O-acyltransferase 1
- AKT1/2
AKT serine/threonine kinases 1 and 2
- ALD
alcoholic liver disease
- ALDH2
aldehyde dehydrogenase 2
- ALK1/5
anaplastic lymphoma receptor tyrosine kinases 1 and 5
- AMOT
angiomotin
- AMPK
AMP-activated protein kinase
- AP-1
activator protein 1
- APOA4
apolipoprotein A4
- ARG1
arginase 1
- ARID1A
AT-rich interaction domain 1A
- ASC
apoptosis-associated speck-like protein containing a CARD
- ASK1
apoptosis signal regulating kinase 1
- ATF
activating transcription factor
- ATG13
autophagy-related gene 13
- ATG14
autophagy-related gene 14
- ATGL
adipose triglyceride lipase
- ATP
adenosine triphosphate
- BA
bile acid
- Bach1
BTB domain and CNC homologue 1
- BCL2
BCL2 apoptosis regulator
- BIRC5
baculoviral IAP repeat containing 5
- CAMKKβ
calcium/calmodulin-dependent protein kinase kinase β
- CASP
caspase
- CASTOR1
cytosolic arginine sensor for mTORC1 subunit 1
- CAT
catalase
- CEBPB
CCAAT enhancer binding protein beta
- CEBPD
CCAAT enhancer binding protein delta
- CCL2/5
C-C motif chemokine ligands 2 and 5
- CCND1
cyclin D1
- CD36
CD36 molecule
- CD44
CD44 molecule
- CD68
CD68 molecule
- CDK6
cyclin-dependent kinase 6
- CDKN1B
cyclin-dependent kinase inhibitor 1B
- C/EBP
CCAAT enhancer binding protein
- CFLAR
CASP8 and FADD apoptosis regulator
- CHOP
CCAAT/enhancer-binding protein homologous protein
- ChoRE
carbohydrate response element
- ChREBP
carbohydrate response element binding protein
- CIDEC
cell death inducing DFFA-like effector C
- CLEC4F
C-type lectin domain family 4 member F
- COL1A1
collagen type I alpha 1 chain
- COL3A1
collagen type III alpha 1 chain
- CPT1
carnitine palmitoyltransferase 1
- CPT1A
carnitine palmitoyltransferase 1A
- CTGF
connective tissue growth factor
- CXCL1/5/10
C-X-C motif chemokine ligands 1, 5, and 10
- CYP2E1
cytochrome P450 family 2 subfamily E member 1
- CYP7A1
cytochrome P450 family 7 subfamily A member 1
- CYP8B1
cytochrome P450 family 8 subfamily B member 1
- CYP27A1
cytochrome P450 family 27 subfamily A member 1
- CYR61
cysteine-rich angiogenic inducer 61
- DAG
diacylglycerol
- DGAT1
diacylglycerol O-acyltransferase 1
- DHCR24
24-dehydrocholesterol reductase
- DNL
de novo lipogenesis
- DNMT3A
DNA methyltransferase 3 alpha
- DPP4
dipeptidyl peptidase 4
- eIF2
eukaryotic translation initiation factor 2
- ER
endoplasmic reticulum
- ERK
extracellular signal-regulated kinase
- EtOH
ethanol
- ETS1
ETS proto-oncogene 1
- EV
extracellular vesicle
- FABP1
fatty acid binding protein 1
- FAO
fatty acid oxidation
- FASN
fatty acid synthase
- FBP1
fructose-bisphosphatase 1
- FGF11
fibroblast growth factor 11
- FGF21
fibroblast growth factor 21
- FGFR1/2
fibroblast growth factor receptors 1 and 2
- FIZZ1
found in inflammatory zone 1
- FN1
fibronectin 1
- Fos
Fos proto-oncogene
- FOXO1/3
forkhead box O 1/3
- FXR
farnesoid X receptor
- FZD
frizzled class receptor
- G6PC
glucose-6-phosphatase catalytic subunit
- G6PD
glucose-6-phosphate dehydrogenase
- GADD34
growth arrest and DNA damage-inducible protein 34
- GATA4/6
GATA binding proteins 4 and 6
- GATOR1/2
GTPase-activating proteins 1/2 toward Rags complex
- GCK
glucokinase
- GlcNAc
N-acetyl-d-glucosamine
- GLI2/3
GLI family zinc finger 2/3
- GLS
glutaminase
- GNA12
guanine nucleotide-binding protein subunit alpha 12
- GPCR
G protein-coupled receptor
- GR
glucocorticoid receptor
- HDAC3
histone deacetylase 3
- HDL
high-density lipoprotein
- HGF
hepatocyte growth factor
- HIF1α
hypoxia inducible factor 1α
- HMGA2
high-mobility group AT-hook 2
- HMGCR
3-hydroxy-3-methylglutaryl-CoA reductase
- HMGCS1/2
3-hydroxy-3-methylglutaryl-CoA synthases 1 and 2
- HMOX1
heme oxygenase 1
- HNRNPA1
heterogeneous nuclear ribonucleoprotein A1
- HSC
hepatic stellate cell
- HSL
hormone-sensitive lipase
- IFNγ
interferon gamma
- IGF1
insulin-like growth factor 1
- IGF1R
insulin-like growth factor 1 receptor
- IGFBP2
IGF binding protein 2
- IHH
Indian hedgehog signaling molecule
- IKBE
NFKB inhibitor epsilon
- IKBKE
inhibitor of nuclear factor kappa B kinase subunit epsilon
- IKK
inhibitor of nuclear factor kappa B kinase
- IL1β
interleukin 1β
- IL6
interleukin 6
- IL1R
interleukin 1 receptor
- IL1RN
interleukin 1 receptor antagonist
- IL6R
interleukin 6 receptor
- INSIG1
insulin-induced gene 1
- IRAK
interleukin 1 receptor-associated kinase
- IRE1α
inositol-requiring enzyme 1 α
- JAK
Janus kinase
- JNK
JUN N-terminal kinase
- JUN
JUN proto-oncogene
- K+
potassium ion
- KC
Kupffer cell
- KIBRA
kidney and brain protein
- KLF4/6
Kruppel-like factors 4 and 6
- LAMP1
lysosomal-associated membrane protein 1
- LATS1/2
large tumor suppressor kinases 1 and 2
- Leu
leucine
- LID
low-glucose inhibitory domain
- LIPG
lipase G
- LKB1
liver kinase B1
- LOXL2
lysyl oxidase-like 2
- LPA
lysophosphatidic acid
- LPL
lipoprotein lipase
- LPS
lipopolysaccharide
- LRP5/6
LDL receptor-related proteins 5 and 6
- LTB4
leukotriene B4
- LXR
liver X receptor
- M2 φ
M2 macrophage
- MAP3K1
mitogen-activated protein kinase kinase kinase 1
- MAP3K3
mitogen-activated protein kinase kinase kinase 3
- MAP4K
mitogen-activated protein kinase kinase kinase kinase
- MAPK
mitogen-activated protein kinase
- MAT1A
methionine adenosyltransferase 1A
- MCPIP
monocyte chemotactic protein-induced protein
- MEK7
MAPK/ERK kinase 7
- MERTK
MER proto-oncogene tyrosine kinase
- Met
methionine
- miR
microRNA
- MKK
mitogen-activated protein kinase kinase
- MMP1/7/14
matrix metallopeptidases 1, 7, and 14
- MOB1A/B
MOB kinase activator 1 A/B
- MRC1
mannose receptor C-type 1
- MST1/2
mammalian STE20-like protein kinases 1 and 2
- MT
metallothionein
- MTF1
metal regulatory transcription factor 1
- mTOR
mechanistic target of rapamycin kinase
- mTORC1/2
mechanistic target of rapamycin kinase complexes 1 and 2
- MYC
MYC proto-oncogene
- MyD88
myeloid differentiation primary response protein 88
- NAD
nicotinamide adenine dinucleotide
- NAFLD
nonalcoholic fatty liver disease
- NASH
nonalcoholic steatohepatitis
- NCOA2
nuclear receptor coactivator 2
- NCOR
nuclear receptor corepressor
- NDRG3
N-Myc downstream-regulated gene 3 protein
- NF2
neurofibromin 2
- NF-κB
nuclear factor kappa B subunit
- NFYC
nuclear transcription factor Y subunit gamma
- NIK
NF-κB-inducing kinase
- NLRP3
NLR family pyrin domain containing 3
- NPC1L1
NPC1-like intracellular cholesterol transporter 1
- NPLCs
nonparenchymal liver cells
- NRF2
nuclear factor erythroid 2-related factor 2
- OGT
O-linked N-acetylglucosamine transferase
- OPN
osteopontin
- P2X7
purinergic receptor P2X 7
- p38 MAPK
p38 mitogen-activated protein kinase
- p300
E1A binding protein p300
- PA
palmitic acid
- PAI-1
plasminogen activator inhibitor 1
- PDGF
platelet-derived growth factor
- PDGFB
platelet-derived growth factor B
- PDGFC
platelet-derived growth factor C
- PDGFRB
platelet-derived growth factor receptor B
- PERK
PRKR-like endoplasmic reticulum kinase
- PGC1α
peroxisome proliferator-activated receptor gamma coactivator 1 alpha
- PI3K
phosphatidylinositol 3-kinase
- PIM3
Pim-3 proto-oncogene serine/threonine kinase
- PKA
protein kinase A
- PKLR
pyruvate kinase L/R
- PKM2
pyruvate kinase M2
- PM
plasma membrane
- PP1C
protein phosphatase 1C
- PP2A
protein phosphatase 2A
- PPAR
peroxisome proliferator-activated receptor
- PRC1
protein regulator of cytokinesis 1
- PTEN
phosphatase and tensin homologue
- PU.1
transcription factor PU.1
- RA
retinoic acid
- RAF
Raf proto-oncogene serine/threonine kinase
- RAG
Ras-related GTP binding protein
- RAS
Ras proto-oncogene GTPase
- Raptor
regulatory associated protein of mTOR complex 1
- REDD1
protein regulated in development and DNA damage response 1
- RELB
RELB proto-oncogene
- RHEB
Ras homologue enriched in brain
- RHOA
Ras homology family member A
- RICTOR
RPTOR independent companion of MTOR complex 2
- RIDD
regulated IRE1-dependent decay
- ROCK1
Rho-associated coiled-coil containing protein kinase 1
- ROS
reactive oxygen species
- RXR
retinoid X receptor
- S6K1
ribosomal protein S6 kinase 1
- SAM
S-adenosylmethionine
- SAMTOR
S-adenosylmethionine sensor upstream of mTORC1
- SAV1
Salvador family WW domain containing protein 1
- SC4MOL
sterol-C4-methyl oxidase
- SCAP
SREBP-cleavage activating protein
- SCD1
stearoyl-CoA desaturase 1
- SESN
sestrin
- sFRP
secreted frizzled related protein
- SHP
small heterodimer partner
- SIRT
sirtuin
- SLC9A3R1
SLC9A3 regulator 1
- SLC27A5
solute carrier family 27 member 5
- SLC43A1
solute carrier family 43 member A1
- SLC51A
solute carrier family 51 subunit alpha
- SMAD
SMAD family member
- SMURF
SMAD-specific E3 ubiquitin protein ligase
- SOCS1
suppressor of cytokine signaling 1
- SOD
superoxide dismutase
- SP3
SP3 transcription factor
- SR-BI
scavenger receptor class B member 1
- SREBP
sterol regulatory element binding protein
- STAT
signal transducer and activator of transcription
- SUMO
sumoylation
- TAK1
transforming growth factor beta-activated kinase 1
- TAZ
transcriptional coactivator with PDZ-binding motif
- TBK1
TANK binding kinase 1
- TCF21
transcription factor 21
- TEAD
TEA domain transcription factor
- TET
ten eleven translocation
- TFE3
transcription factor binding to IGHM enhancer 3
- TFEB
transcription factor EB
- TG
triglyceride
- TGFβ
transforming growth factor β
- TGFBR1
transforming growth factor beta receptor 1
- TIMP1
TIMP metallopeptidase inhibitor 1
- TIRAP
TIR domain containing adaptor protein
- TLR4
toll-like receptor 4
- TNF
tumor necrosis factor
- TNFR
tumor necrosis factor receptor
- TRADD
tumor necrosis factor receptor type 1-associated death domain protein
- TRAF
tumor necrosis factor receptor-associated factor
- TRIM4
tripartite motif containing 4
- TSC
tuberous sclerosis complex subunit
- TUBA1B
tubulin alpha 1b
- TXNIP
thioredoxin interacting protein
- ULK1
Unc-51-like autophagy activating kinase 1
- UPR
unfolded protein response
- UVRAG
UV radiation resistance associated
- VGLL4
vestigial-like family member 4
- VHL
von Hippel–Lindau tumor suppressor
- VLDL
very low-density lipoprotein
- VLDLR
very low-density lipoprotein receptor
- WNT
Wnt family member
- WT
wild type
- WWTR1
WW domain containing transcription regulator 1
- XBP1
X-box binding protein 1
- XBP1s
X-box binding protein 1 spliced
- XBP1u
X-box binding protein 1 unspliced
- YAP
yes1-associated transcriptional regulator
- ZEB2
zinc finger E-box binding homeobox 2
Authors' Contributions
X.C.D. contributed to the concept development, literature searches, illustration preparation, and article writing and revision. M.H. contributed to the article draft preparation, especially the signal transduction section. H.G.K. contributed to the article draft preparation, especially the section of sirtuins, microRNAs, and DNA methylation. K.C. contributed to the article draft preparation, especially the section of Transcriptional Regulation of Hepatic Steatosis, Inflammation, and Fibrosis.
Author Disclosure Statement
No competing financial interests exist.
Funding Information
This work was supported by the U.S. National Institute of Diabetes and Digestive and Kidney Diseases of the National Institutes of Health under award numbers R01DK120689 (Xiaocheng Charlie Dong) and R01DK121925 (Xiaocheng Charlie Dong) and the Indiana Clinical and Translational Sciences Institute funded, in part, by award number UL1TR002529 from the National Center for Advancing Translational Sciences of the National Institutes of Health. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
References
- 1. Adachi M, Osawa Y, Uchinami H, Kitamura T, Accili D, and Brenner DA. The forkhead transcription factor FoxO1 regulates proliferation and transdifferentiation of hepatic stellate cells. Gastroenterology 132: 1434–1446, 2007. [DOI] [PubMed] [Google Scholar]
- 2. Ahrens M, Ammerpohl O, von Schonfels W, Kolarova J, Bens S, Itzel T, Teufel A, Herrmann A, Brosch M, Hinrichsen H, Erhart W, Egberts J, Sipos B, Schreiber S, Hasler R, Stickel F, Becker T, Krawczak M, Rocken C, Siebert R, Schafmayer C, and Hampe J. DNA methylation analysis in nonalcoholic fatty liver disease suggests distinct disease-specific and remodeling signatures after bariatric surgery. Cell Metab 18: 296–302, 2013. [DOI] [PubMed] [Google Scholar]
- 3. Alsamman S, Christenson SA, Yu A, Ayad NME, Mooring MS, Segal JM, Hu JK, Schaub JR, Ho SS, Rao V, Marlow MM, Turner SM, Sedki M, Pantano L, Ghoshal S, Ferreira DDS, Ma HY, Duwaerts CC, Espanol-Suner R, Wei L, Newcomb B, Mileva I, Canals D, Hannun YA, Chung RT, Mattis AN, Fuchs BC, Tager AM, Yimlamai D, Weaver VM, Mullen AC, Sheppard D, and Chen JY. Targeting acid ceramidase inhibits YAP/TAZ signaling to reduce fibrosis in mice. Sci Transl Med 12: eaay878, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Anakk S, Bhosale M, Schmidt VA, Johnson RL, Finegold MJ, and Moore DD. Bile acids activate YAP to promote liver carcinogenesis. Cell Rep 5: 1060–1069, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Ao X, Ding W, Zhang Y, Ding D, and Liu Y. TCF21: a critical transcription factor in health and cancer. J Mol Med (Berl) 98: 1055–1068, 2020. [DOI] [PubMed] [Google Scholar]
- 6. Asrani SK, Mellinger J, Arab JP, and Shah VH. Reducing the global burden of alcohol-associated liver disease: a blueprint for action. Hepatology 73: 2039–2050, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Aylon Y, Gershoni A, Rotkopf R, Biton IE, Porat Z, Koh AP, Sun X, Lee Y, Fiel MI, Hoshida Y, Friedman SL, Johnson RL, and Oren M. The LATS2 tumor suppressor inhibits SREBP and suppresses hepatic cholesterol accumulation. Genes Dev 30: 786–797, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Babuta M, Furi I, Bala S, Bukong TN, Lowe P, Catalano D, Calenda C, Kodys K, and Szabo G. Dysregulated autophagy and lysosome function are linked to exosome production by micro-RNA 155 in alcoholic liver disease. Hepatology 70: 2123–2141, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Bala S, Csak T, Kodys K, Catalano D, Ambade A, Furi I, Lowe P, Cho Y, Iracheta-Vellve A, and Szabo G. Alcohol-induced miR-155 and HDAC11 inhibit negative regulators of the TLR4 pathway and lead to increased LPS responsiveness of Kupffer cells in alcoholic liver disease. J Leukoc Biol 102: 487–498, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Bala S, Csak T, Saha B, Zatsiorsky J, Kodys K, Catalano D, Satishchandran A, and Szabo G. The pro-inflammatory effects of miR-155 promote liver fibrosis and alcohol-induced steatohepatitis. J Hepatol 64: 1378–1387, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Bang IH, Kwon OK, Hao L, Park D, Chung MJ, Oh BC, Lee S, Bae EJ, and Park BH. Deacetylation of XBP1s by sirtuin 6 confers resistance to ER stress-induced hepatic steatosis. Exp Mol Med 51: 1–11, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Barcena-Varela M, Paish H, Alvarez L, Uriarte I, Latasa MU, Santamaria E, Recalde M, Garate M, Claveria A, Colyn L, Arechederra M, Iraburu MJ, Milkiewicz M, Milkiewicz P, Sangro B, Robinson SM, French J, Pardo-Saganta A, Oyarzabal J, Prosper F, Rombouts K, Oakley F, Mann J, Berasain C, Avila MA, and Fernandez-Barrena MG. Epigenetic mechanisms and metabolic reprogramming in fibrogenesis: dual targeting of G9a and DNMT1 for the inhibition of liver fibrosis. Gut 70: 388–400, 2021. [DOI] [PubMed] [Google Scholar]
- 13. Baumeier C, Saussenthaler S, Kammel A, Jahnert M, Schluter L, Hesse D, Canouil M, Lobbens S, Caiazzo R, Raverdy V, Pattou F, Nilsson E, Pihlajamaki J, Ling C, Froguel P, Schurmann A, and Schwenk RW. Hepatic DPP4 DNA methylation associates with fatty liver. Diabetes 66: 25–35, 2017. [DOI] [PubMed] [Google Scholar]
- 14. Benhamed F, Denechaud PD, Lemoine M, Robichon C, Moldes M, Bertrand-Michel J, Ratziu V, Serfaty L, Housset C, Capeau J, Girard J, Guillou H, and Postic C. The lipogenic transcription factor ChREBP dissociates hepatic steatosis from insulin resistance in mice and humans. J Clin Invest 122: 2176–2194, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Beraza N, Malato Y, Vander Borght S, Liedtke C, Wasmuth HE, Dreano M, de Vos R, Roskams T, and Trautwein C. Pharmacological IKK2 inhibition blocks liver steatosis and initiation of non-alcoholic steatohepatitis. Gut 57: 655–663, 2008. [DOI] [PubMed] [Google Scholar]
- 16. Calvente CJ, Tameda M, Johnson CD, Del Pilar H, Lin YC, Adronikou N, De Mollerat Du Jeu X, Llorente C, Boyer J, and Feldstein AE. Neutrophils contribute to spontaneous resolution of liver inflammation and fibrosis via microRNA-223. J Clin Invest 129: 4091–4109, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Canto C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, Elliott PJ, Puigserver P, and Auwerx J. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 458: 1056–1060, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Caron S, Huaman Samanez C, Dehondt H, Ploton M, Briand O, Lien F, Dorchies E, Dumont J, Postic C, Cariou B, Lefebvre P, Staels B. and Farnesoid X receptor inhibits the transcriptional activity of carbohydrate response element binding protein in human hepatocytes. Mol Cell Biol 33: 2202–2211, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Catrysse L and van Loo G. Inflammation and the metabolic syndrome: the tissue-specific functions of nF-kappaB. Trends Cell Biol 27: 417–429, 2017. [DOI] [PubMed] [Google Scholar]
- 20. Caviglia JM, Yan J, Jang MK, Gwak GY, Affo S, Yu L, Olinga P, Friedman RA, Chen X, and Schwabe RF. MicroRNA-21 and Dicer are dispensable for hepatic stellate cell activation and the development of liver fibrosis. Hepatology 67: 2414–2429, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Cha JY and Repa JJ. The liver X receptor (LXR) and hepatic lipogenesis. The carbohydrate-response element-binding protein is a target gene of LXR. J Biol Chem 282: 743–751, 2007. [DOI] [PubMed] [Google Scholar]
- 22. Chai C, Cox B, Yaish D, Gross D, Rosenberg N, Amblard F, Shemuelian Z, Gefen M, Korach A, Tirosh O, Lanton T, Link H, Tam J, Permyakova A, Ozhan G, Citrin J, Liao H, Tannous M, Hahn M, Axelrod J, Arretxe E, Alonso C, Martinez-Arranz I, Betes PO, Safadi R, Salhab A, Amer J, Tber Z, Mengshetti S, Giladi H, Schinazi RF, and Galun E. Agonist of RORA attenuates nonalcoholic fatty liver progression in mice via up-regulation of microRNA 122. Gastroenterology 159: 999–1014.e9, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Chatani N, Kamada Y, Kizu T, Ogura S, Furuta K, Egawa M, Hamano M, Ezaki H, Kiso S, Shimono A, Ouchi N, Yoshida Y, and Takehara T. Secreted frizzled-related protein 5 (Sfrp5) decreases hepatic stellate cell activation and liver fibrosis. Liver Int 35: 2017–2026, 2015. [DOI] [PubMed] [Google Scholar]
- 24. Chen H, Shen F, Sherban A, Nocon A, Li Y, Wang H, Xu MJ, Rui X, Han J, Jiang B, Lee D, Li N, Keyhani-Nejad F, Fan JG, Liu F, Kamat A, Musi N, Guarente L, Pacher P, Gao B, and Zang M. DEP domain-containing mTOR-interacting protein suppresses lipogenesis and ameliorates hepatic steatosis and acute-on-chronic liver injury in alcoholic liver disease. Hepatology 68: 496–514, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Chen X, Zhang F, Gong Q, Cui A, Zhuo S, Hu Z, Han Y, Gao J, Sun Y, Liu Z, Yang Z, Le Y, Gao X, Dong LQ, Gao X, and Li Y. Hepatic ATF6 increases fatty acid oxidation to attenuate hepatic steatosis in mice through peroxisome proliferator-activated receptor alpha. Diabetes 65: 1904–1915, 2016. [DOI] [PubMed] [Google Scholar]
- 26. Cheng J, Liu C, Hu K, Greenberg A, Wu D, Ausman LM, McBurney MW, and Wang XD. Ablation of systemic SIRT1 activity promotes nonalcoholic fatty liver disease by affecting liver-mesenteric adipose tissue fatty acid mobilization. Biochim Biophys Acta Mol Basis Dis 1863: 2783–2790, 2017. [DOI] [PubMed] [Google Scholar]
- 27. Ciesielska A, Matyjek M, and Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol Life Sci 78: 1233–1261, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Collin de l'Hortet A, Takeishi K, Guzman-Lepe J, Morita K, Achreja A, Popovic B, Wang Y, Handa K, Mittal A, Meurs N, Zhu Z, Weinberg F, Salomon M, Fox IJ, Deng CX, Nagrath D, and Soto-Gutierrez A. Generation of human fatty livers using custom-engineered induced pluripotent stem cells with modifiable SIRT1 metabolism. Cell Metab 30: 385–401 e9, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Cotter TG and Rinella M. Nonalcoholic fatty liver disease 2020: the state of the disease. Gastroenterology 158: 1851–1864, 2020. [DOI] [PubMed] [Google Scholar]
- 30. Crisafulli C and Cuzzocrea S. The role of endogenous and exogenous ligands for the peroxisome proliferator-activated receptor alpha (PPAR-alpha) in the regulation of inflammation in macrophages. Shock 32: 62–73, 2009. [DOI] [PubMed] [Google Scholar]
- 31. Csak T, Bala S, Lippai D, Kodys K, Catalano D, Iracheta-Vellve A, and Szabo G. MicroRNA-155 deficiency attenuates liver steatosis and fibrosis without reducing inflammation in a mouse model of steatohepatitis. PLoS One 10: e0129251, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Czuwara-Ladykowska J, Sementchenko VI, Watson DK, and Trojanowska M. Ets1 is an effector of the transforming growth factor beta (TGF-beta) signaling pathway and an antagonist of the profibrotic effects of TGF-beta. J Biol Chem 277: 20399–20408, 2002. [DOI] [PubMed] [Google Scholar]
- 33. da Silva Morais A, Abarca-Quinones J, Guigas B, Viollet B, Starkel P, Horsmans Y, and Leclercq IA. Development of hepatic fibrosis occurs normally in AMPK-deficient mice. Clin Sci (Lond) 118: 411–420, 2009. [DOI] [PubMed] [Google Scholar]
- 34. Dela Pena A, Leclercq I, Field J, George J, Jones B, and Farrell G. NF-kappaB activation, rather than TNF, mediates hepatic inflammation in a murine dietary model of steatohepatitis. Gastroenterology 129: 1663–1674, 2005. [DOI] [PubMed] [Google Scholar]
- 35. Delgado I, Carrasco M, Cano E, Carmona R, García-Carbonero R, Marín-Gómez LM, Soria B, Martín F, Cano DA, Muñoz-Chápuli R, and Rojas A. GATA4 loss in the septum transversum mesenchyme promotes liver fibrosis in mice. Hepatology 59: 2358–2370, 2014. [DOI] [PubMed] [Google Scholar]
- 36. Deng X, Zhang W, I OS, Williams JB, Dong Q, Park EA, Raghow R, Unterman TG, and Elam MB. FoxO1 inhibits sterol regulatory element-binding protein-1c (SREBP-1c) gene expression via transcription factors Sp1 and SREBP-1c. J Biol Chem 287: 20132–20143, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Dentin R, Benhamed F, Hainault I, Fauveau V, Foufelle F, Dyck JR, Girard J, and Postic C. Liver-specific inhibition of ChREBP improves hepatic steatosis and insulin resistance in ob/ob mice. Diabetes 55: 2159–2170, 2006. [DOI] [PubMed] [Google Scholar]
- 38. Devchand PR, Keller H, Peters JM, Vazquez M, Gonzalez FJ, and Wahli W. The PPARalpha-leukotriene B4 pathway to inflammation control. Nature 384: 39–43, 1996. [DOI] [PubMed] [Google Scholar]
- 39. Dong XC. FOXO transcription factors in non-alcoholic fatty liver disease. Liver Res 1: 168–173, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Dong XC, Copps KD, Guo S, Li Y, Kollipara R, DePinho RA, and White MF. Inactivation of hepatic Foxo1 by insulin signaling is required for adaptive nutrient homeostasis and endocrine growth regulation. Cell Metab 8: 65–76, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Driskill JH and Pan D. The hippo pathway in liver homeostasis and pathophysiology. Annu Rev Pathol 16: 299–322, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Du K, Hyun J, Premont RT, Choi SS, Michelotti GA, Swiderska-Syn M, Dalton GD, Thelen E, Rizi BS, Jung Y, and Diehl AM. Hedgehog-YAP signaling pathway regulates glutaminolysis to control activation of hepatic stellate cells. Gastroenterology 154: 1465–1479.e13, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Dupont S, Morsut L, Aragona M, Enzo E, Giulitti S, Cordenonsi M, Zanconato F, Le Digabel J, Forcato M, Bicciato S, Elvassore N, and Piccolo S. Role of YAP/TAZ in mechanotransduction. Nature 474: 179–183, 2011. [DOI] [PubMed] [Google Scholar]
- 44. Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, and Manning BD. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol Cell 39: 171–183, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA, Mair W, Vasquez DS, Joshi A, Gwinn DM, Taylor R, Asara JM, Fitzpatrick J, Dillin A, Viollet B, Kundu M, Hansen M, and Shaw RJ. Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science 331: 456–461, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Egnatchik RA, Leamy AK, Jacobson DA, Shiota M, and Young JD. ER calcium release promotes mitochondrial dysfunction and hepatic cell lipotoxicity in response to palmitate overload. Mol Metab 3: 544–553, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Elhanati S, Ben-Hamo R, Kanfi Y, Varvak A, Glazz R, Lerrer B, Efroni S, and Cohen HY. Reciprocal regulation between SIRT6 and miR-122 controls liver metabolism and predicts hepatocarcinoma prognosis. Cell Rep 14: 234–242, 2016. [DOI] [PubMed] [Google Scholar]
- 48. Fabregat I, Moreno-Caceres J, Sanchez A, Dooley S, Dewidar B, Giannelli G, Ten Dijke P, and Consortium I-L. TGF-beta signalling and liver disease. FEBS J 283: 2219–2232, 2016. [DOI] [PubMed] [Google Scholar]
- 49. Fan W, Morinaga H, Kim JJ, Bae E, Spann NJ, Heinz S, Glass CK, and Olefsky JM. FoxO1 regulates Tlr4 inflammatory pathway signalling in macrophages. EMBO J 29: 4223–4236, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Feige JN, Lagouge M, Canto C, Strehle A, Houten SM, Milne JC, Lambert PD, Mataki C, Elliott PJ, and Auwerx J. Specific SIRT1 activation mimics low energy levels and protects against diet-induced metabolic disorders by enhancing fat oxidation. Cell Metab 8: 347–358, 2008. [DOI] [PubMed] [Google Scholar]
- 51. Fisher FM, Kim M, Doridot L, Cunniff JC, Parker TS, Levine DM, Hellerstein MK, Hudgins LC, Maratos-Flier E, and Herman MA. A critical role for ChREBP-mediated FGF21 secretion in hepatic fructose metabolism. Mol Metab 6: 14–21, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Gao J, Wei B, de Assuncao TM, Liu Z, Hu X, Ibrahim S, Cooper SA, Cao S, Shah VH, and Kostallari E. Hepatic stellate cell autophagy inhibits extracellular vesicle release to attenuate liver fibrosis. J Hepatol 73: 1144–1154, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Garcia D, Hellberg K, Chaix A, Wallace M, Herzig S, Badur MG, Lin T, Shokhirev MN, Pinto AFM, Ross DS, Saghatelian A, Panda S, Dow LE, Metallo CM, and Shaw RJ. Genetic liver-specific AMPK activation protects against diet-induced obesity and NAFLD. Cell Rep 26: 192–208.e6, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Gavrilova O, Haluzik M, Matsusue K, Cutson JJ, Johnson L, Dietz KR, Nicol CJ, Vinson C, Gonzalez FJ, and Reitman ML. Liver peroxisome proliferator-activated receptor gamma contributes to hepatic steatosis, triglyceride clearance, and regulation of body fat mass. J Biol Chem 278: 34268–34276, 2003. [DOI] [PubMed] [Google Scholar]
- 55. Goedeke L, Salerno A, Ramirez CM, Guo L, Allen RM, Yin X, Langley SR, Esau C, Wanschel A, Fisher EA, Suarez Y, Baldan A, Mayr M, and Fernandez-Hernando C. Long-term therapeutic silencing of miR-33 increases circulating triglyceride levels and hepatic lipid accumulation in mice. EMBO Mol Med 6: 1133–1141, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Gonzalez A, Hall MN, Lin SC, and Hardie DG. AMPK and TOR: the Yin and Yang of cellular nutrient sensing and growth control. Cell Metab 31: 472–492, 2020. [DOI] [PubMed] [Google Scholar]
- 57. Gonzalez A, Huerta-Salgado C, Orozco-Aguilar J, Aguirre F, Tacchi F, Simon F, and Cabello-Verrugio C. Role of oxidative stress in hepatic and extrahepatic dysfunctions during nonalcoholic fatty liver disease (NAFLD). Oxid Med Cell Longev 2020: 1617805, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Grohmann M, Wiede F, Dodd GT, Gurzov EN, Ooi GJ, Butt T, Rasmiena AA, Kaur S, Gulati T, Goh PK, Treloar AE, Archer S, Brown WA, Muller M, Watt MJ, Ohara O, McLean CA, and Tiganis T. Obesity drives STAT-1-dependent NASH and STAT-3-dependent HCC. Cell 175: 1289–1306.e20, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Haeusler RA, Hartil K, Vaitheesvaran B, Arrieta-Cruz I, Knight CM, Cook JR, Kammoun HL, Febbraio MA, Gutierrez-Juarez R, Kurland IJ, and Accili D. Integrated control of hepatic lipogenesis versus glucose production requires FoxO transcription factors. Nat Commun 5: 5190, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Hagiwara A, Cornu M, Cybulski N, Polak P, Betz C, Trapani F, Terracciano L, Heim MH, Ruegg MA, and Hall MN. Hepatic mTORC2 activates glycolysis and lipogenesis through Akt, glucokinase, and SREBP1c. Cell Metab 15: 725–738, 2012. [DOI] [PubMed] [Google Scholar]
- 61. He Y, Hwang S, Cai Y, Kim SJ, Xu M, Yang D, Guillot A, Feng D, Seo W, Hou X, and Gao B. MicroRNA-223 ameliorates nonalcoholic steatohepatitis and cancer by targeting multiple inflammatory and oncogenic genes in hepatocytes. Hepatology 70: 1150–1167, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. He Y, Rodrigues RM, Wang X, Seo W, Ma J, Hwang S, Fu Y, Trojnar E, Matyas C, Zhao S, Ren R, Feng D, Pacher P, Kunos G, and Gao B. Neutrophil-to-hepatocyte communication via LDLR-dependent miR-223-enriched extracellular vesicle transfer ameliorates nonalcoholic steatohepatitis. J Clin Invest 131: e141513, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Hernandez-Gea V, Ghiassi-Nejad Z, Rozenfeld R, Gordon R, Fiel MI, Yue Z, Czaja MJ, and Friedman SL. Autophagy releases lipid that promotes fibrogenesis by activated hepatic stellate cells in mice and in human tissues. Gastroenterology 142: 938–946, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Hernandez-Gea V, Hilscher M, Rozenfeld R, Lim MP, Nieto N, Werner S, Devi LA, and Friedman SL. Endoplasmic reticulum stress induces fibrogenic activity in hepatic stellate cells through autophagy. J Hepatol 59: 98–104, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Hess J, Angel P, and Schorpp-Kistner M. AP-1 subunits: quarrel and harmony among siblings. J Cell Sci 117: 5965–5973, 2004. [DOI] [PubMed] [Google Scholar]
- 66. Hsu SH, Wang B, Kota J, Yu J, Costinean S, Kutay H, Yu L, Bai S, La Perle K, Chivukula RR, Mao H, Wei M, Clark KR, Mendell JR, Caligiuri MA, Jacob ST, Mendell JT, and Ghoshal K. Essential metabolic, anti-inflammatory, and anti-tumorigenic functions of miR-122 in liver. J Clin Invest 122: 2871–2883, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Huang M, Kim HG, Zhong X, Dong C, Zhang B, Fang Z, Zhang Y, Lu X, Saxena R, Liu Y, Zhang C, Liangpunsakul S, and Dong XC. Sestrin 3 protects against diet-induced nonalcoholic steatohepatitis in mice through suppression of transforming growth factor beta signal transduction. Hepatology 71: 76–92, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Hung YH, Kanke M, Kurtz CL, Cubitt RL, Bunaciu RP, Zhou L, White PJ, Vickers KC, Hussain MM, Li X, and Sethupathy P. MiR-29 regulates de novo lipogenesis in the liver and circulating triglyceride levels in a Sirt1-dependent manner. Front Physiol 10: 1367, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Hyun J and Jung Y. DNA methylation in nonalcoholic fatty liver disease. Int J Mol Sci 21: 8138, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Iizuka K, Bruick RK, Liang G, Horton JD, and Uyeda K. Deficiency of carbohydrate response element-binding protein (ChREBP) reduces lipogenesis as well as glycolysis. Proc Natl Acad Sci U S A 101: 7281–7286, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Inoue Y, Itoh Y, Abe K, Okamoto T, Daitoku H, Fukamizu A, Onozaki K, and Hayashi H. Smad3 is acetylated by p300/CBP to regulate its transactivation activity. Oncogene 26: 500–508, 2007. [DOI] [PubMed] [Google Scholar]
- 72. Jegal KH, Park SM, Cho SS, Byun SH, Ku SK, Kim SC, Ki SH, and Cho IJ. Activating transcription factor 6-dependent sestrin 2 induction ameliorates ER stress-mediated liver injury. Biochim Biophys Acta Mol Cell Res 1864: 1295–1307, 2017. [DOI] [PubMed] [Google Scholar]
- 73. Jeong SH, Kim HB, Kim MC, Lee JM, Lee JH, Kim JH, Kim JW, Park WY, Kim SY, Kim JB, Kim H, Kim JM, Choi HS, and Lim DS. Hippo-mediated suppression of IRS2/AKT signaling prevents hepatic steatosis and liver cancer. J Clin Invest 128: 1010–1025, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Jiang F, Parsons CJ, and Stefanovic B. Gene expression profile of quiescent and activated rat hepatic stellate cells implicates Wnt signaling pathway in activation. J Hepatol 45: 401–409, 2006. [DOI] [PubMed] [Google Scholar]
- 75. Jiang H, Westerterp M, Wang C, Zhu Y, and Ai D. Macrophage mTORC1 disruption reduces inflammation and insulin resistance in obese mice. Diabetologia 57: 2393–2404, 2014. [DOI] [PubMed] [Google Scholar]
- 76. Jiang HY, Wek SA, McGrath BC, Scheuner D, Kaufman RJ, Cavener DR, and Wek RC. Phosphorylation of the alpha subunit of eukaryotic initiation factor 2 is required for activation of NF-kappaB in response to diverse cellular stresses. Mol Cell Biol 23: 5651–5663, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Ka SO, Bang IH, Bae EJ, and Park BH. Hepatocyte-specific sirtuin 6 deletion predisposes to nonalcoholic steatohepatitis by up-regulation of Bach1, an Nrf2 repressor. FASEB J 31: 3999–4010, 2017. [DOI] [PubMed] [Google Scholar]
- 78. Kamada Y, Yoshino K, Kondo C, Kawamata T, Oshiro N, Yonezawa K, and Ohsumi Y. Tor directly controls the Atg1 kinase complex to regulate autophagy. Mol Cell Biol 30: 1049–1058, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Kapoor N, Niu J, Saad Y, Kumar S, Sirakova T, Becerra E, Li X, and Kolattukudy PE. Transcription factors STAT6 and KLF4 implement macrophage polarization via the dual catalytic powers of MCPIP. J Immunol 194: 6011–6023, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Kennedy MA, Barrera GC, Nakamura K, Baldan A, Tarr P, Fishbein MC, Frank J, Francone OL, and Edwards PA. ABCG1 has a critical role in mediating cholesterol efflux to HDL and preventing cellular lipid accumulation. Cell Metab 1: 121–131, 2005. [DOI] [PubMed] [Google Scholar]
- 81. Kersten S, Seydoux J, Peters JM, Gonzalez FJ, Desvergne B, and Wahli W. Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting. J Clin Invest 103: 1489–1498, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Kim HG, Huang M, Xin Y, Zhang Y, Zhang X, Wang G, Liu S, Wan J, Ahmadi AR, Sun Z, Liangpunsakul S, Xiong X, and Dong XC. The epigenetic regulator SIRT6 protects the liver from alcohol-induced tissue injury by reducing oxidative stress in mice. J Hepatol 71: 960–969, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Kim HS, Xiao C, Wang RH, Lahusen T, Xu X, Vassilopoulos A, Vazquez-Ortiz G, Jeong WI, Park O, Ki SH, Gao B, and Deng CX. Hepatic-specific disruption of SIRT6 in mice results in fatty liver formation due to enhanced glycolysis and triglyceride synthesis. Cell Metab 12: 224–236, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Kim J, Kundu M, Viollet B, and Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13: 132–141, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Kim KH, Choi S, Zhou Y, Kim EY, Lee JM, Saha PK, Anakk S, and Moore DD. Hepatic FXR/SHP axis modulates systemic glucose and fatty acid homeostasis in aged mice. Hepatology 66: 498–509, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Kim KM, Han CY, Kim JY, Cho SS, Kim YS, Koo JH, Lee JM, Lim SC, Kang KW, Kim JS, Hwang SJ, Ki SH, and Kim SG. Galpha12 overexpression induced by miR-16 dysregulation contributes to liver fibrosis by promoting autophagy in hepatic stellate cells. J Hepatol 68: 493–504, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Kim YC, Seok S, Zhang Y, Ma J, Kong B, Guo G, Kemper B, and Kemper JK. Intestinal FGF15/19 physiologically repress hepatic lipogenesis in the late fed-state by activating SHP and DNMT3A. Nat Commun 11: 5969, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Kirchgessner TG, Sleph P, Ostrowski J, Lupisella J, Ryan CS, Liu X, Fernando G, Grimm D, Shipkova P, Zhang R, Garcia R, Zhu J, He A, Malone H, Martin R, Behnia K, Wang Z, Barrett YC, Garmise RJ, Yuan L, Zhang J, Gandhi MD, Wastall P, Li T, Du S, Salvador L, Mohan R, Cantor GH, Kick E, Lee J, and Frost RJ. Beneficial and adverse effects of an LXR agonist on human lipid and lipoprotein metabolism and circulating neutrophils. Cell Metab 24: 223–233, 2016. [DOI] [PubMed] [Google Scholar]
- 89. Knittel T, Kobold D, Dudas J, Saile B, and Ramadori G. Role of the Ets-1 transcription factor during activation of rat hepatic stellate cells in culture. Am J Pathol 155: 1841–1848, 1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Knorr J, Wree A, Tacke F, and Feldstein AE. The NLRP3 inflammasome in alcoholic and nonalcoholic steatohepatitis. Semin Liver Dis 40: 298–306, 2020. [DOI] [PubMed] [Google Scholar]
- 91. Kong B, Wang L, Chiang JY, Zhang Y, Klaassen CD, and Guo GL. Mechanism of tissue-specific farnesoid X receptor in suppressing the expression of genes in bile-acid synthesis in mice. Hepatology 56: 1034–1043, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Koo JH, Lee HJ, Kim W, and Kim SG. Endoplasmic reticulum stress in hepatic stellate cells promotes liver fibrosis via PERK-mediated degradation of HNRNPA1 and up-regulation of SMAD2. Gastroenterology 150: 181–193.e8, 2016. [DOI] [PubMed] [Google Scholar]
- 93. Lai SS, Fu X, Cheng Q, Yu ZH, Jiang EZ, Zhao DD, Yu DC, Qiu YD, Gao X, Ju HX, Wang W, Jiang Q, Zhu MS, Li CJ, and Xue B. HSC-specific knockdown of GGPPS alleviated CCl4-induced chronic liver fibrosis through mediating RhoA/Rock pathway. Am J Transl Res 11: 2382–2392, 2019. [PMC free article] [PubMed] [Google Scholar]
- 94. Latella G, Vetuschi A, Sferra R, Catitti V, D'Angelo A, Zanninelli G, Flanders KC, and Gaudio E. Targeted disruption of Smad3 confers resistance to the development of dimethylnitrosamine-induced hepatic fibrosis in mice. Liver Int 29: 997–1009, 2009. [DOI] [PubMed] [Google Scholar]
- 95. Lebeaupin C, Vallee D, Rousseau D, Patouraux S, Bonnafous S, Adam G, Luciano F, Luci C, Anty R, Iannelli A, Marchetti S, Kroemer G, Lacas-Gervais S, Tran A, Gual P, and Bailly-Maitre B. Bax inhibitor-1 protects from nonalcoholic steatohepatitis by limiting inositol-requiring enzyme 1 alpha signaling in mice. Hepatology 68: 515–532, 2018. [DOI] [PubMed] [Google Scholar]
- 96. Lee AH, Scapa EF, Cohen DE, and Glimcher LH. Regulation of hepatic lipogenesis by the transcription factor XBP1. Science 320: 1492–1496, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Lee GY, Kim NH, Zhao ZS, Cha BS, and Kim YS. Peroxisomal-proliferator-activated receptor alpha activates transcription of the rat hepatic malonyl-CoA decarboxylase gene: a key regulation of malonyl-CoA level. Biochem J 378: 983–990, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Lee JH, Budanov AV, Talukdar S, Park EJ, Park HL, Park HW, Bandyopadhyay G, Li N, Aghajan M, Jang I, Wolfe AM, Perkins GA, Ellisman MH, Bier E, Scadeng M, Foretz M, Viollet B, Olefsky J, and Karin M. Maintenance of metabolic homeostasis by Sestrin2 and Sestrin3. Cell Metab 16: 311–321, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Lee MK, Pardoux C, Hall MC, Lee PS, Warburton D, Qing J, Smith SM, and Derynck R. TGF-beta activates Erk MAP kinase signalling through direct phosphorylation of ShcA. EMBO J 26: 3957–3967, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Lee MY, Choi R, Kim HM, Cho EJ, Kim BH, Choi YS, Naowaboot J, Lee EY, Yang YC, Shin JY, Shin YG, and Chung CH. Peroxisome proliferator-activated receptor delta agonist attenuates hepatic steatosis by anti-inflammatory mechanism. Exp Mol Med 44: 578–585, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Lee Y, Ka SO, Cha HN, Chae YN, Kim MK, Park SY, Bae EJ, and Park BH. Myeloid Sirtuin 6 deficiency causes insulin resistance in high-fat diet-fed mice by eliciting macrophage polarization toward an M1 phenotype. Diabetes 66: 2659–2668, 2017. [DOI] [PubMed] [Google Scholar]
- 102. Lerner AG, Upton JP, Praveen PV, Ghosh R, Nakagawa Y, Igbaria A, Shen S, Nguyen V, Backes BJ, Heiman M, Heintz N, Greengard P, Hui S, Tang Q, Trusina A, Oakes SA, and Papa FR. IRE1alpha induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell Metab 16: 250–264, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Li G, Hao W, and Hu W. Transcription factor PU.1 and immune cell differentiation (Review). Int J Mol Med 46: 1943–1950, 2020. [DOI] [PubMed] [Google Scholar]
- 104. Li H, Meng Q, Xiao F, Chen S, Du Y, Yu J, Wang C, and Guo F. ATF4 deficiency protects mice from high-carbohydrate-diet-induced liver steatosis. Biochem J 438: 283–289, 2011. [DOI] [PubMed] [Google Scholar]
- 105. Li M, He Y, Zhou Z, Ramirez T, Gao Y, Gao Y, Ross RA, Cao H, Cai Y, Xu M, Feng D, Zhang P, Liangpunsakul S, and Gao B. MicroRNA-223 ameliorates alcoholic liver injury by inhibiting the IL-6-p47(phox)-oxidative stress pathway in neutrophils. Gut 66: 705–715, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Li MV, Chang B, Imamura M, Poungvarin N, and Chan L. Glucose-dependent transcriptional regulation by an evolutionarily conserved glucose-sensing module. Diabetes 55: 1179–1189, 2006. [DOI] [PubMed] [Google Scholar]
- 107. Li Y, Wong K, Giles A, Jiang J, Lee JW, Adams AC, Kharitonenkov A, Yang Q, Gao B, Guarente L, and Zang M. Hepatic SIRT1 attenuates hepatic steatosis and controls energy balance in mice by inducing fibroblast growth factor 21. Gastroenterology 146: 539–549 e7, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Li Y, Xu S, Mihaylova MM, Zheng B, Hou X, Jiang B, Park O, Luo Z, Lefai E, Shyy JY, Gao B, Wierzbicki M, Verbeuren TJ, Shaw RJ, Cohen RA, and Zang M. AMPK phosphorylates and inhibits SREBP activity to attenuate hepatic steatosis and atherosclerosis in diet-induced insulin-resistant mice. Cell Metab 13: 376–388, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Liao X, Sharma N, Kapadia F, Zhou G, Lu Y, Hong H, Paruchuri K, Mahabeleshwar GH, Dalmas E, Venteclef N, Flask CA, Kim J, Doreian BW, Lu KQ, Kaestner KH, Hamik A, Clement K, and Jain MK. Kruppel-like factor 4 regulates macrophage polarization. J Clin Invest 121: 2736–2749, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Lim JY, Oh MA, Kim WH, Sohn HY, and Park SI. AMP-activated protein kinase inhibits TGF-beta-induced fibrogenic responses of hepatic stellate cells by targeting transcriptional coactivator p300. J Cell Physiol 227: 1081–1089, 2012. [DOI] [PubMed] [Google Scholar]
- 111. Lin HY, Wang FS, Yang YL, and Huang YH. MicroRNA-29a Suppresses CD36 to ameliorate high fat diet-induced steatohepatitis and liver fibrosis in mice. Cells 8: 1298, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Lin Y, Ding D, Huang Q, Liu Q, Lu H, Lu Y, Chi Y, Sun X, Ye G, Zhu H, Wei J, and Dong S. Downregulation of miR-192 causes hepatic steatosis and lipid accumulation by inducing SREBF1: novel mechanism for bisphenol A-triggered non-alcoholic fatty liver disease. Biochim Biophys Acta Mol Cell Biol Lipids 1862: 869–882, 2017. [DOI] [PubMed] [Google Scholar]
- 113. Liu C, Gaca MD, Swenson ES, Vellucci VF, Reiss M, and Wells RG. Smads 2 and 3 are differentially activated by transforming growth factor-beta (TGF-beta) in quiescent and activated hepatic stellate cells. Constitutive nuclear localization of Smads in activated cells is TGF-beta-independent. J Biol Chem 278: 11721–11728, 2003. [DOI] [PubMed] [Google Scholar]
- 114. Liu GY and Sabatini DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol 21: 183–203, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Liu Q, Yu J, Wang L, Tang Y, Zhou Q, Ji S, Wang Y, Santos L, Haeusler RA, Que J, Rajbhandari P, Lei X, Valenti L, Pajvani UB, Qin J, and Qiang L. Inhibition of PU.1 ameliorates metabolic dysfunction and non-alcoholic steatohepatitis. J Hepatol 73: 361–370, 2020. [DOI] [PubMed] [Google Scholar]
- 116. Liu S, Hatano B, Zhao M, Yen CC, Kang K, Reilly SM, Gangl MR, Gorgun C, Balschi JA, Ntambi JM, and Lee CH. Role of peroxisome proliferator-activated receptor {delta}/{beta} in hepatic metabolic regulation. J Biol Chem 286: 1237–1247, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Liu X, Xu J, Rosenthal S, Zhang LJ, McCubbin R, Meshgin N, Shang L, Koyama Y, Ma HY, Sharma S, Heinz S, Glass CK, Benner C, Brenner DA, and Kisseleva T. Identification of lineage-specific transcription factors that prevent activation of hepatic stellate cells and promote fibrosis resolution. Gastroenterology 158: 1728–1744.e14, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Liu XL, Pan Q, Cao HX, Xin FZ, Zhao ZH, Yang RX, Zeng J, Zhou H, and Fan JG. Lipotoxic hepatocyte-derived exosomal microRNA 192–195p activates macrophages through Rictor/Akt/Forkhead box transcription factor O1 signaling in nonalcoholic fatty liver disease. Hepatology 72: 454–469, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Long JK, Dai W, Zheng YW, and Zhao SP. miR-122 promotes hepatic lipogenesis via inhibiting the LKB1/AMPK pathway by targeting Sirt1 in non-alcoholic fatty liver disease. Mol Med 25: 26, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Lu XY, Shi XJ, Hu A, Wang JQ, Ding Y, Jiang W, Sun M, Zhao X, Luo J, Qi W, and Song BL. Feeding induces cholesterol biosynthesis via the mTORC1-USP20-HMGCR axis. Nature 588: 479–484, 2020. [DOI] [PubMed] [Google Scholar]
- 121. Luedde T and Schwabe RF. NF-kappaB in the liver—linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 8: 108–118, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Ma J, Cao H, Rodrigues RM, Xu M, Ren T, He Y, Hwang S, Feng D, Ren R, Yang P, Liangpunsakul S, Sun J, and Gao B. Chronic-plus-binge alcohol intake induces production of proinflammatory mtDNA-enriched extracellular vesicles and steatohepatitis via ASK1/p38MAPKalpha-dependent mechanisms. JCI Insight 5: e13696, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Manmadhan S and Ehmer U. Hippo signaling in the liver—a long and ever-expanding story. Front Cell Dev Biol 7: 33, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Mannaerts I, Leite SB, Verhulst S, Claerhout S, Eysackers N, Thoen LF, Hoorens A, Reynaert H, Halder G, and van Grunsven LA. The Hippo pathway effector YAP controls mouse hepatic stellate cell activation. J Hepatol 63: 679–688, 2015. [DOI] [PubMed] [Google Scholar]
- 125. Mansouri A, Gattolliat CH, and Asselah T. Mitochondrial dysfunction and signaling in chronic liver diseases. Gastroenterology 155: 629–647, 2018. [DOI] [PubMed] [Google Scholar]
- 126. Marrone G, De Chiara F, Bottcher K, Levi A, Dhar D, Longato L, Mazza G, Zhang Z, Marrali M, Fernandez-Iglesias A, Hall A, Luong TV, Viollet B, Pinzani M, and Rombouts K. The adenosine monophosphate-activated protein kinase-vacuolar adenosine triphosphatase-pH axis: a key regulator of the profibrogenic phenotype of human hepatic stellate cells. Hepatology 68: 1140–1153, 2018. [DOI] [PubMed] [Google Scholar]
- 127. Martin K, Pritchett J, Llewellyn J, Mullan AF, Athwal VS, Dobie R, Harvey E, Zeef L, Farrow S, Streuli C, Henderson NC, Friedman SL, Hanley NA, and Piper Hanley K. PAK proteins and YAP-1 signalling downstream of integrin beta-1 in myofibroblasts promote liver fibrosis. Nat Commun 7: 12502, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Masri S, Rigor P, Cervantes M, Ceglia N, Sebastian C, Xiao C, Roqueta-Rivera M, Deng C, Osborne TF, Mostoslavsky R, Baldi P, and Sassone-Corsi P. Partitioning circadian transcription by SIRT6 leads to segregated control of cellular metabolism. Cell 158: 659–672, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Matsumoto Y, Itami S, Kuroda M, Yoshizato K, Kawada N, and Murakami Y. MiR-29a assists in preventing the activation of human stellate cells and promotes recovery from liver fibrosis in mice. Mol Ther 24: 1848–1859, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. McMullen PD, Bhattacharya S, Woods CG, Sun B, Yarborough K, Ross SM, Miller ME, McBride MT, LeCluyse EL, Clewell RA, and Andersen ME. A map of the PPARalpha transcription regulatory network for primary human hepatocytes. Chem Biol Interact 209: 14–24, 2014. [DOI] [PubMed] [Google Scholar]
- 131. Min HK, Kapoor A, Fuchs M, Mirshahi F, Zhou H, Maher J, Kellum J, Warnick R, Contos MJ, and Sanyal AJ. Increased hepatic synthesis and dysregulation of cholesterol metabolism is associated with the severity of nonalcoholic fatty liver disease. Cell Metab 15: 665–674, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Moon YA, Liang G, Xie X, Frank-Kamenetsky M, Fitzgerald K, Koteliansky V, Brown MS, Goldstein JL, and Horton JD. The Scap/SREBP pathway is essential for developing diabetic fatty liver and carbohydrate-induced hypertriglyceridemia in animals. Cell Metab 15: 240–246, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Mooring M, Fowl BH, Lum SZC, Liu Y, Yao K, Softic S, Kirchner R, Bernstein A, Singhi AD, Jay DG, Kahn CR, Camargo FD, and Yimlamai D. Hepatocyte stress increases expression of Yes-associated protein and transcriptional coactivator with PDZ-binding motif in hepatocytes to promote parenchymal inflammation and fibrosis. Hepatology 71: 1813–1830, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Morán-Salvador E, López-Parra M, García-Alonso V, Titos E, Martínez-Clemente M, González-Périz A, López-Vicario C, Barak Y, Arroyo V, and Clària J. Role for PPARγ in obesity-induced hepatic steatosis as determined by hepatocyte- and macrophage-specific conditional knockouts. FASEB J 25: 2538–2550, 2011. [DOI] [PubMed] [Google Scholar]
- 135. Moran-Salvador E, Titos E, Rius B, Gonzalez-Periz A, Garcia-Alonso V, Lopez-Vicario C, Miquel R, Barak Y, Arroyo V, and Claria J. Cell-specific PPARgamma deficiency establishes anti-inflammatory and anti-fibrogenic properties for this nuclear receptor in non-parenchymal liver cells. J Hepatol 59: 1045–1053, 2013. [DOI] [PubMed] [Google Scholar]
- 136. Munday MR, Campbell DG, Carling D, and Hardie DG. Identification by amino acid sequencing of three major regulatory phosphorylation sites on rat acetyl-CoA carboxylase. Eur J Biochem 175: 331–338, 1988. [DOI] [PubMed] [Google Scholar]
- 137. Murphy SK, Yang H, Moylan CA, Pang H, Dellinger A, Abdelmalek MF, Garrett ME, Ashley-Koch A, Suzuki A, Tillmann HL, Hauser MA, and Diehl AM. Relationship between methylome and transcriptome in patients with nonalcoholic fatty liver disease. Gastroenterology 145: 1076–1087, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Musso G, Gambino R, and Cassader M. Cholesterol metabolism and the pathogenesis of non-alcoholic steatohepatitis. Prog Lipid Res 52: 175–191, 2013. [DOI] [PubMed] [Google Scholar]
- 139. Mwinyi J, Bostrom AE, Pisanu C, Murphy SK, Erhart W, Schafmayer C, Hampe J, Moylan C, and Schioth HB. NAFLD is associated with methylation shifts with relevance for the expression of genes involved in lipoprotein particle composition. Biochim Biophys Acta Mol Cell Biol Lipids 1862: 314–323, 2017. [DOI] [PubMed] [Google Scholar]
- 140. Myung SJ, Yoon JH, Gwak GY, Kim W, Lee JH, Kim KM, Shin CS, Jang JJ, Lee SH, Lee SM, and Lee HS. Wnt signaling enhances the activation and survival of human hepatic stellate cells. FEBS Lett 581: 2954–2958, 2007. [DOI] [PubMed] [Google Scholar]
- 141. Nagaya T, Tanaka N, Suzuki T, Sano K, Horiuchi A, Komatsu M, Nakajima T, Nishizawa T, Joshita S, Umemura T, Ichijo T, Matsumoto A, Yoshizawa K, Nakayama J, Tanaka E, and Aoyama T. Down-regulation of SREBP-1c is associated with the development of burned-out NASH. J Hepatol 53: 724–731, 2010. [DOI] [PubMed] [Google Scholar]
- 142. Naiman S, Huynh FK, Gil R, Glick Y, Shahar Y, Touitou N, Nahum L, Avivi MY, Roichman A, Kanfi Y, Gertler AA, Doniger T, Ilkayeva OR, Abramovich I, Yaron O, Lerrer B, Gottlieb E, Harris RA, Gerber D, Hirschey MD, and Cohen HY. SIRT6 promotes hepatic beta-oxidation via activation of PPARalpha. Cell Rep 29: 4127–4143.e8, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, Gerszten RE, and Naar AM. MicroRNA-33 and the SREBP host genes cooperate to control cholesterol homeostasis. Science 328: 1566–1569, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Nakano Y, Kamiya A, Sumiyoshi H, Tsuruya K, Kagawa T, and Inagaki Y. A Deactivation factor of fibrogenic hepatic stellate cells induces regression of liver fibrosis in mice. Hepatology 71: 1437–1452, 2020. [DOI] [PubMed] [Google Scholar]
- 145. Ni HM, Chao X, Yang H, Deng F, Wang S, Bai Q, Qian H, Cui Y, Cui W, Shi Y, Zong WX, Wang Z, Yang L, and Ding WX. Dual roles of mammalian target of rapamycin in regulating liver injury and tumorigenesis in autophagy-defective mouse liver. Hepatology 70: 2142–2155, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, Mukundan L, Red Eagle A, Vats D, Brombacher F, Ferrante AW, and Chawla A. Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature 447: 1116–1120, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Odegaard JI, Ricardo-Gonzalez RR, Red Eagle A, Vats D, Morel CR, Goforth MH, Subramanian V, Mukundan L, Ferrante AW, and Chawla A. Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance. Cell Metab 7: 496–507, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Ortega-Prieto P and Postic C. Carbohydrate sensing through the transcription factor ChREBP. Front Genet 10: 472, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Oyadomari S, Harding HP, Zhang Y, Oyadomari M, and Ron D. Dephosphorylation of translation initiation factor 2alpha enhances glucose tolerance and attenuates hepatosteatosis in mice. Cell Metab 7: 520–532, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Ozaki I, Zhao G, Mizuta T, Ogawa Y, Hara T, Kajihara S, Hisatomi A, Sakai T, and Yamamoto K. Hepatocyte growth factor induces collagenase (matrix metalloproteinase-1) via the transcription factor Ets-1 in human hepatic stellate cell line. J Hepatol 36: 169–178, 2002. [DOI] [PubMed] [Google Scholar]
- 151. Page A, Paoli P, Moran Salvador E, White S, French J, and Mann J. Hepatic stellate cell transdifferentiation involves genome-wide remodeling of the DNA methylation landscape. J Hepatol 64: 661–673, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Pan Q, Guo CJ, Xu QY, Wang JZ, Li H, and Fang CH. miR-16 integrates signal pathways in myofibroblasts: determinant of cell fate necessary for fibrosis resolution. Cell Death Dis 11: 639, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Pan X, Zhang Y, Kim H-G, Liangpunsakul S, and Dong XC. FOXO transcription factors protect against the diet-induced fatty liver disease. Sci Rep 7: 44597, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Park HW, Park H, Ro SH, Jang I, Semple IA, Kim DN, Kim M, Nam M, Zhang D, Yin L, and Lee JH. Hepatoprotective role of Sestrin2 against chronic ER stress. Nat Commun 5: 4233, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Park S-J, Sohn H-Y, Yoon J, and Park SI. Down-regulation of FoxO-dependent c-FLIP expression mediates TRAIL-induced apoptosis in activated hepatic stellate cells. Cell Signall 21: 1495–1503, 2009. [DOI] [PubMed] [Google Scholar]
- 156. Patsenker E, Schneider V, Ledermann M, Saegesser H, Dorn C, Hellerbrand C, and Stickel F. Potent antifibrotic activity of mTOR inhibitors sirolimus and everolimus but not of cyclosporine A and tacrolimus in experimental liver fibrosis. J Hepatol 55: 388–398, 2011. [DOI] [PubMed] [Google Scholar]
- 157. Pawlak M, Lefebvre P, and Staels B. Molecular mechanism of PPARalpha action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol 62: 720–733, 2015. [DOI] [PubMed] [Google Scholar]
- 158. Peet DJ, Turley SD, Ma W, Janowski BA, Lobaccaro JM, Hammer RE, and Mangelsdorf DJ. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell 93: 693–704, 1998. [DOI] [PubMed] [Google Scholar]
- 159. Pettinelli P and Videla LA. Up-regulation of PPAR-gamma mRNA expression in the liver of obese patients: an additional reinforcing lipogenic mechanism to SREBP-1c induction. J Clin Endocrinol Metab 96: 1424–1430, 2011. [DOI] [PubMed] [Google Scholar]
- 160. Pineda Torra I, Claudel T, Duval C, Kosykh V, Fruchart JC, and Staels B. Bile acids induce the expression of the human peroxisome proliferator-activated receptor alpha gene via activation of the farnesoid X receptor. Mol Endocrinol 17: 259–272, 2003. [DOI] [PubMed] [Google Scholar]
- 161. Ponugoti B, Kim DH, Xiao Z, Smith Z, Miao J, Zang M, Wu SY, Chiang CM, Veenstra TD, and Kemper JK. SIRT1 deacetylates and inhibits SREBP-1C activity in regulation of hepatic lipid metabolism. J Biol Chem 285: 33959–33970, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, and Li X. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab 9: 327–338, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Rayner KJ, Esau CC, Hussain FN, McDaniel AL, Marshall SM, van Gils JM, Ray TD, Sheedy FJ, Goedeke L, Liu X, Khatsenko OG, Kaimal V, Lees CJ, Fernandez-Hernando C, Fisher EA, Temel RE, and Moore KJ. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature 478: 404–407, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Rayner KJ, Suarez Y, Davalos A, Parathath S, Fitzgerald ML, Tamehiro N, Fisher EA, Moore KJ, and Fernandez-Hernando C. MiR-33 contributes to the regulation of cholesterol homeostasis. Science 328: 1570–1573, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Repa JJ, Liang G, Ou J, Bashmakov Y, Lobaccaro JM, Shimomura I, Shan B, Brown MS, Goldstein JL, and Mangelsdorf DJ. Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRalpha and LXRbeta. Genes Dev 14: 2819–2830, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Reyes-Gordillo K, Shah R, Arellanes-Robledo J, Cheng Y, Ibrahim J, and Tuma PL. Akt1 and Akt2 isoforms play distinct roles in regulating the development of inflammation and fibrosis associated with alcoholic liver disease. Cells 8: 1337, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Ricote M, Li AC, Willson TM, Kelly CJ, and Glass CK. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 391: 79–82, 1998. [DOI] [PubMed] [Google Scholar]
- 168. Roderburg C, Urban GW, Bettermann K, Vucur M, Zimmermann H, Schmidt S, Janssen J, Koppe C, Knolle P, Castoldi M, Tacke F, Trautwein C, and Luedde T. Micro-RNA profiling reveals a role for miR-29 in human and murine liver fibrosis. Hepatology 53: 209–218, 2011. [DOI] [PubMed] [Google Scholar]
- 169. Roy S, Benz F, Alder J, Bantel H, Janssen J, Vucur M, Gautheron J, Schneider A, Schuller F, Loosen S, Luedde M, Koch A, Tacke F, Luedde T, and Trautwein C, Roderburg C. Down-regulation of miR-192-5p protects from oxidative stress-induced acute liver injury. Clin Sci (Lond) 130: 1197–1207, 2016. [DOI] [PubMed] [Google Scholar]
- 170. Rutkowski DT, Wu J, Back SH, Callaghan MU, Ferris SP, Iqbal J, Clark R, Miao H, Hassler JR, Fornek J, Katze MG, Hussain MM, Song B, Swathirajan J, Wang J, Yau GD, and Kaufman RJ. UPR pathways combine to prevent hepatic steatosis caused by ER stress-mediated suppression of transcriptional master regulators. Dev Cell 15: 829–840, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Saha B, Bala S, Hosseini N, Kodys K, and Szabo G. Kruppel-like factor 4 is a transcriptional regulator of M1/M2 macrophage polarization in alcoholic liver disease. J Leukoc Biol 97: 963–973, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Sanderson LM, Boekschoten MV, Desvergne B, Muller M, and Kersten S. Transcriptional profiling reveals divergent roles of PPARalpha and PPARbeta/delta in regulation of gene expression in mouse liver. Physiol Genomics 41: 42–52, 2010. [DOI] [PubMed] [Google Scholar]
- 173. Satapati S, Kucejova B, Duarte JA, Fletcher JA, Reynolds L, Sunny NE, He T, Nair LA, Livingston KA, Fu X, Merritt ME, Sherry AD, Malloy CR, Shelton JM, Lambert J, Parks EJ, Corbin I, Magnuson MA, Browning JD, and Burgess SC. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J Clin Invest 125: 4447–4462, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Satishchandran A, Ambade A, Rao S, Hsueh YC, Iracheta-Vellve A, Tornai D, Lowe P, Gyongyosi B, Li J, Catalano D, Zhong L, Kodys K, Xie J, Bala S, Gao G, and Szabo G. MicroRNA 122, regulated by GRLH2, protects livers of mice and patients from ethanol-induced liver disease. Gastroenterology 154: 238–252.e7, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Schmidt-Arras D and Rose-John S. IL-6 pathway in the liver: from physiopathology to therapy. J Hepatol 64: 1403–1415, 2016. [DOI] [PubMed] [Google Scholar]
- 176. Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, Heyman RA, Briggs M, Deeb S, Staels B, and Auwerx J. PPARalpha and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene. EMBO J 15: 5336–5348, 1996. [PMC free article] [PubMed] [Google Scholar]
- 177. Schug TT, Xu Q, Gao H, Peres-da-Silva A, Draper DW, Fessler MB, Purushotham A, and Li X. Myeloid deletion of SIRT1 induces inflammatory signaling in response to environmental stress. Mol Cell Biol 30: 4712–4721, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Schulien I, Hockenjos B, Schmitt-Graeff A, Perdekamp MG, Follo M, Thimme R, and Hasselblatt P. The transcription factor c-Jun/AP-1 promotes liver fibrosis during non-alcoholic steatohepatitis by regulating Osteopontin expression. Cell Death Differ 26: 1688–1699, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Schuster S, Cabrera D, Arrese M, and Feldstein AE. Triggering and resolution of inflammation in NASH. Nat Rev Gastroenterol Hepatol 15: 349–364, 2018. [DOI] [PubMed] [Google Scholar]
- 180. Senokuchi T, Liang CP, Seimon TA, Han S, Matsumoto M, Banks AS, Paik JH, DePinho RA, Accili D, Tabas I, and Tall AR. Forkhead transcription factors (FoxOs) promote apoptosis of insulin-resistant macrophages during cholesterol-induced endoplasmic reticulum stress. Diabetes 57: 2967–2976, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Seo E, Park EJ, Joe Y, Kang S, Kim MS, Hong SH, Park MK, Kim DK, Koh H, and Lee HJ. Overexpression of AMPKalpha1 ameliorates fatty liver in hyperlipidemic diabetic rats. Korean J Physiol Pharmacol 13: 449–454, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Shan L, Ding Y, Fu Y, Zhou L, Dong X, Chen S, Wu H, Nai W, Zheng H, Xu W, Bai X, Jia C, and Dai M. mTOR overactivation in mesenchymal cells aggravates CCl4- induced liver fibrosis. Sci Rep 6: 36037, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Sharifnia T, Antoun J, Verriere TG, Suarez G, Wattacheril J, Wilson KT, Peek RM Jr., Abumrad NN, and Flynn CR.. Hepatic TLR4 signaling in obese NAFLD. Am J Physiol Gastrointest Liver Physiol 309: G270–G278, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Shimano H, Horton JD, Shimomura I, Hammer RE, Brown MS, and Goldstein JL. Isoform 1c of sterol regulatory element binding protein is less active than isoform 1a in livers of transgenic mice and in cultured cells. J Clin Invest 99: 846–854, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Shimano H and Sato R. SREBP-regulated lipid metabolism: convergent physiology—divergent pathophysiology. Nat Rev Endocrinol 13: 710–730, 2017. [DOI] [PubMed] [Google Scholar]
- 186. Sinal CJ, Tohkin M, Miyata M, Ward JM, Lambert G, and Gonzalez FJ. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 102: 731–744, 2000. [DOI] [PubMed] [Google Scholar]
- 187. So JS, Hur KY, Tarrio M, Ruda V, Frank-Kamenetsky M, Fitzgerald K, Koteliansky V, Lichtman AH, Iwawaki T, Glimcher LH, and Lee AH. Silencing of lipid metabolism genes through IRE1alpha-mediated mRNA decay lowers plasma lipids in mice. Cell Metab 16: 487–499, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Sookoian S, Rosselli MS, Gemma C, Burgueno AL, Fernandez Gianotti T, Castano GO, and Pirola CJ. Epigenetic regulation of insulin resistance in nonalcoholic fatty liver disease: impact of liver methylation of the peroxisome proliferator-activated receptor gamma coactivator 1alpha promoter. Hepatology 52: 1992–2000, 2010. [DOI] [PubMed] [Google Scholar]
- 189. Sorrentino G, Ruggeri N, Specchia V, Cordenonsi M, Mano M, Dupont S, Manfrin A, Ingallina E, Sommaggio R, Piazza S, Rosato A, Piccolo S, and Del Sal G. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat Cell Biol 16: 357–366, 2014. [DOI] [PubMed] [Google Scholar]
- 190. Tang LY, Heller M, Meng Z, Yu LR, Tang Y, Zhou M, and Zhang YE. Transforming growth factor-beta (TGF-beta) directly activates the JAK1-STAT3 axis to induce hepatic fibrosis in coordination with the SMAD pathway. J Biol Chem 292: 4302–4312, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Tao R, Wei D, Gao H, Liu Y, DePinho RA, and Dong XC. Hepatic FoxOs regulate lipid metabolism via modulation of expression of the nicotinamide phosphoribosyltransferase gene. J Biol Chem 286: 14681–14690, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Tao R, Xiong X, DePinho RA, Deng CX, and Dong XC. Hepatic SREBP-2 and cholesterol biosynthesis are regulated by FoxO3 and Sirt6. J Lipid Res 54: 2745–2753, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Tao R, Xiong X, Liangpunsakul S, and Dong XC. Sestrin 3 protein enhances hepatic insulin sensitivity by direct activation of the mTORC2-Akt signaling. Diabetes 64: 1211–1223, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Tian Y, Arai E, Makiuchi S, Tsuda N, Kuramoto J, Ohara K, Takahashi Y, Ito N, Ojima H, Hiraoka N, Gotoh M, Yoshida T, and Kanai Y. Aberrant DNA methylation results in altered gene expression in non-alcoholic steatohepatitis-related hepatocellular carcinomas. J Cancer Res Clin Oncol 146: 2461–2477, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Tsai WC, Hsu SD, Hsu CS, Lai TC, Chen SJ, Shen R, Huang Y, Chen HC, Lee CH, Tsai TF, Hsu MT, Wu JC, Huang HD, Shiao MS, Hsiao M, and Tsou AP. MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J Clin Invest 122: 2884–2897, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Umemura A, Park EJ, Taniguchi K, Lee JH, Shalapour S, Valasek MA, Aghajan M, Nakagawa H, Seki E, Hall MN, and Karin M. Liver damage, inflammation, and enhanced tumorigenesis after persistent mTORC1 inhibition. Cell Metab 20: 133–144, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Usui M, Yamaguchi S, Tanji Y, Tominaga R, Ishigaki Y, Fukumoto M, Katagiri H, Mori K, Oka Y, and Ishihara H. Atf6alpha-null mice are glucose intolerant due to pancreatic beta-cell failure on a high-fat diet but partially resistant to diet-induced insulin resistance. Metabolism 61: 1118–1128, 2012. [DOI] [PubMed] [Google Scholar]
- 198. Van Nostrand JL, Hellberg K, Luo EC, Van Nostrand EL, Dayn A, Yu J, Shokhirev MN, Dayn Y, Yeo GW, and Shaw RJ. AMPK regulation of Raptor and TSC2 mediate metformin effects on transcriptional control of anabolism and inflammation. Genes Dev 34: 1330–1344, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Vander Ark A, Cao J, and Li X. TGF-beta receptors: in and beyond TGF-beta signaling. Cell Signal 52: 112–120, 2018. [DOI] [PubMed] [Google Scholar]
- 200. Venkateswaran A, Laffitte BA, Joseph SB, Mak PA, Wilpitz DC, Edwards PA, and Tontonoz P. Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXR alpha. Proc Natl Acad Sci U S A 97: 12097–12102, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Vickers KC, Landstreet SR, Levin MG, Shoucri BM, Toth CL, Taylor RC, Palmisano BT, Tabet F, Cui HL, Rye KA, Sethupathy P, and Remaley AT. MicroRNA-223 coordinates cholesterol homeostasis. Proc Natl Acad Sci U S A 111: 14518–14523, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Vila L, Elias I, Roca C, Ribera A, Ferre T, Casellas A, Lage R, Franckhauser S, and Bosch F. AAV8-mediated Sirt1 gene transfer to the liver prevents high carbohydrate diet-induced nonalcoholic fatty liver disease. Mol Ther Methods Clin Dev 1: 14039, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Virtue A, Johnson C, Lopez-Pastrana J, Shao Y, Fu H, Li X, Li YF, Yin Y, Mai J, Rizzo V, Tordoff M, Bagi Z, Shan H, Jiang X, Wang H, and Yang XF. MicroRNA-155 deficiency leads to decreased atherosclerosis, increased white adipose tissue obesity, and non-alcoholic fatty liver disease: a novel mouse model of obesity paradox. J Biol Chem 292: 1267–1287, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Wang B and Tontonoz P. Liver X receptors in lipid signalling and membrane homeostasis. Nat Rev Endocrinol 14: 452–463, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Wang K, Zhou W, Cai Q, Cheng J, Cai R, and Xing R. SUMOylation of KLF4 promotes IL-4 induced macrophage M2 polarization. Cell Cycle 16: 374–381, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Wang S, Chen Z, Lam V, Han J, Hassler J, Finck BN, Davidson NO, and Kaufman RJ. IRE1alpha-XBP1s induces PDI expression to increase MTP activity for hepatic VLDL assembly and lipid homeostasis. Cell Metab 16: 473–486, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Wang X, Cai B, Yang X, Sonubi OO, Zheng Z, Ramakrishnan R, Shi H, Valenti L, Pajvani UB, Sandhu J, Infante RE, Radhakrishnan A, Covey DF, Guan KL, Buck J, Levin LR, Tontonoz P, Schwabe RF, and Tabas I. Cholesterol stabilizes TAZ in hepatocytes to promote experimental non-alcoholic steatohepatitis. Cell Metab 31: 969–986.e7, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Wang X, He Y, Mackowiak B, and Gao B. MicroRNAs as regulators, biomarkers and therapeutic targets in liver diseases. Gut 70: 784–795, 2021. [DOI] [PubMed] [Google Scholar]
- 209. Wang X, Sommerfeld MR, Jahn-Hofmann K, Cai B, Filliol A, Remotti HE, Schwabe RF, Kannt A, and Tabas I. A Therapeutic silencing RNA targeting hepatocyte taz prevents and reverses fibrosis in nonalcoholic steatohepatitis in mice. Hepatol Commun 3: 1221–1234, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Wang X, Zheng Z, Caviglia JM, Corey KE, Herfel TM, Cai B, Masia R, Chung RT, Lefkowitch JH, Schwabe RF, and Tabas I. Hepatocyte TAZ/WWTR1 promotes inflammation and fibrosis in nonalcoholic steatohepatitis. Cell Metab 24: 848–862, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Watanabe M, Houten SM, Wang L, Moschetta A, Mangelsdorf DJ, Heyman RA, Moore DD, and Auwerx J. Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c. J Clin Invest 113: 1408–1418, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Winkler M, Staniczek T, Kurschner SW, Schmid CD, Schonhaber H, Cordero J, Kessler L, Mathes A, Sticht C, Nessling M, Uvarovskii A, Anders S, Zhang XJ, von Figura G, Hartmann D, Mogler C, Dobreva G, Schledzewski K, Geraud C, Koch PS, and Goerdt S. Endothelial GATA4 controls liver fibrosis and regeneration by preventing a pathogenic switch in angiocrine signaling. J Hepatol 74: 380–393, 2021. [DOI] [PubMed] [Google Scholar]
- 213. Wong SW, Kwon MJ, Choi AM, Kim HP, Nakahira K, and Hwang DH. Fatty acids modulate Toll-like receptor 4 activation through regulation of receptor dimerization and recruitment into lipid rafts in a reactive oxygen species-dependent manner. J Biol Chem 284: 27384–27392, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Wu H, Ng R, Chen X, Steer CJ, and Song G. MicroRNA-21 is a potential link between non-alcoholic fatty liver disease and hepatocellular carcinoma via modulation of the HBP1-p53-Srebp1c pathway. Gut 65: 1850–1860, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Xiao C, Wang RH, Lahusen TJ, Park O, Bertola A, Maruyama T, Reynolds D, Chen Q, Xu X, Young HA, Chen WJ, Gao B, and Deng CX. Progression of chronic liver inflammation and fibrosis driven by activation of c-JUN signaling in Sirt6 mutant mice. J Biol Chem 287: 41903–41913, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Xiong X, Tao R, DePinho RA, and Dong XC. The autophagy-related gene 14 (Atg14) is regulated by forkhead box O transcription factors and circadian rhythms and plays a critical role in hepatic autophagy and lipid metabolism. J Biol Chem 287: 39107–39114, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Yahagi N, Shimano H, Hasty AH, Matsuzaka T, Ide T, Yoshikawa T, Amemiya-Kudo M, Tomita S, Okazaki H, Tamura Y, Iizuka Y, Ohashi K, Osuga J, Harada K, Gotoda T, Nagai R, Ishibashi S, and Yamada N. Absence of sterol regulatory element-binding protein-1 (SREBP-1) ameliorates fatty livers but not obesity or insulin resistance in Lep(ob)/Lep(ob) mice. J Biol Chem 277: 19353–19357, 2002. [DOI] [PubMed] [Google Scholar]
- 218. Yamashita M, Fatyol K, Jin C, Wang X, Liu Z, and Zhang YE. TRAF6 mediates Smad-independent activation of JNK and p38 by TGF-beta. Mol Cell 31: 918–924, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Yan J, Tung HC, Li S, Niu Y, Garbacz WG, Lu P, Bi Y, Li Y, He J, Xu M, Ren S, Monga SP, Schwabe RF, Yang D, and Xie W. Aryl hydrocarbon receptor signaling prevents activation of hepatic stellate cells and liver fibrogenesis in mice. Gastroenterology 157: 793–806.e14, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Yang AQ, Li D, Chi L, and Ye XS. Validation, identification, and biological consequences of the site-specific O-GlcNAcylation dynamics of carbohydrate-responsive element-binding protein (ChREBP). Mol Cell Proteomics 16: 1233–1243, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Yecies JL, Zhang HH, Menon S, Liu S, Yecies D, Lipovsky AI, Gorgun C, Kwiatkowski DJ, Hotamisligil GS, Lee CH, and Manning BD. Akt stimulates hepatic SREBP1c and lipogenesis through parallel mTORC1-dependent and independent pathways. Cell Metab 14: 21–32, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, and Mayo MW. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J 23: 2369–2380, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Yin H, Hu M, Liang X, Ajmo JM, Li X, Bataller R, Odena G, Stevens SM Jr., and You M. Deletion of SIRT1 from hepatocytes in mice disrupts lipin-1 signaling and aggravates alcoholic fatty liver. Gastroenterology 146: 801–811, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Yoshida K and Matsuzaki K. Differential regulation of TGF-beta/Smad signaling in hepatic stellate cells between acute and chronic liver injuries. Front Physiol 3: 53, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. You M, Matsumoto M, Pacold CM, Cho WK, and Crabb DW. The role of AMP-activated protein kinase in the action of ethanol in the liver. Gastroenterology 127: 1798–1808, 2004. [DOI] [PubMed] [Google Scholar]
- 226. Yu FX, Zhao B, Panupinthu N, Jewell JL, Lian I, Wang LH, Zhao J, Yuan H, Tumaneng K, Li H, Fu XD, Mills GB, and Guan KL. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 150: 780–791, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Yu L, Wang L, Yi H, and Wu X. LRP6-CRISPR prevents activation of hepatic stellate cells and liver fibrogenesis in rats. Am J Transl Res 12: 397–408, 2020. [PMC free article] [PubMed] [Google Scholar]
- 228. Yu L, York J, von Bergmann K, Lutjohann D, Cohen JC, and Hobbs HH. Stimulation of cholesterol excretion by the liver X receptor agonist requires ATP-binding cassette transporters G5 and G8. J Biol Chem 278: 15565–15570, 2003. [DOI] [PubMed] [Google Scholar]
- 229. Zarei M, Aguilar-Recarte D, Palomer X, and Vazquez-Carrera M. Revealing the role of peroxisome proliferator-activated receptor beta/delta in nonalcoholic fatty liver disease. Metabolism 114: 154342, 2021. [DOI] [PubMed] [Google Scholar]
- 230. Zeng L, Lu M, Mori K, Luo S, Lee AS, Zhu Y, and Shyy JY. ATF6 modulates SREBP2-mediated lipogenesis. EMBO J 23: 950–958, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Zeybel M, Hardy T, Robinson SM, Fox C, Anstee QM, Ness T, Masson S, Mathers JC, French J, White S, and Mann J. Differential DNA methylation of genes involved in fibrosis progression in non-alcoholic fatty liver disease and alcoholic liver disease. Clin Epigenetics 7: 25, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Zhang D, Tong X, VanDommelen K, Gupta N, Stamper K, Brady GF, Meng Z, Lin J, Rui L, Omary MB, and Yin L. Lipogenic transcription factor ChREBP mediates fructose-induced metabolic adaptations to prevent hepatotoxicity. J Clin Invest 127: 2855–2867, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Zhang J, Li Y, Liu Q, Huang Y, Li R, Wu T, Zhang Z, Zhou J, Huang H, Tang Q, Huang C, Zhao Y, Zhang G, Jiang W, Mo L, Zhang J, Xie W, and He J. Sirt6 alleviated liver fibrosis by deacetylating conserved lysine 54 on Smad2 in hepatic stellate cells. Hepatology 73: 1140–1157, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Zhang K, Li L, Qi Y, Zhu X, Gan B, DePinho RA, Averitt T, and Guo S. Hepatic suppression of Foxo1 and Foxo3 causes hypoglycemia and hyperlipidemia in mice. Endocrinology 153: 631–646, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Zhang K, Wang S, Malhotra J, Hassler JR, Back SH, Wang G, Chang L, Xu W, Miao H, Leonardi R, Chen YE, Jackowski S, and Kaufman RJ. The unfolded protein response transducer IRE1alpha prevents ER stress-induced hepatic steatosis. EMBO J 30: 1357–1375, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Zhang W, Bu SY, Mashek MT, I OS, Sibai Z, Khan SA, Ilkayeva O, Newgard CB, Mashek DG, and Unterman TG. Integrated regulation of hepatic lipid and glucose metabolism by adipose triacylglycerol lipase and FoxO proteins. Cell Rep 15: 349–359, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Zhang W, Patil S, Chauhan B, Guo S, Powell DR, Le J, Klotsas A, Matika R, Xiao X, Franks R, Heidenreich KA, Sajan MP, Farese RV, Stolz DB, Tso P, Koo SH, Montminy M, and Unterman TG. FoxO1 regulates multiple metabolic pathways in the liver: effects on gluconeogenic, glycolytic, and lipogenic gene expression. J Biol Chem 281: 10105–10117, 2006. [DOI] [PubMed] [Google Scholar]
- 238. Zhao P, Sun X, Chaggan C, Liao Z, In Wong K, He F, Singh S, Loomba R, Karin M, Witztum JL, and Saltiel AR. An AMPK-caspase-6 axis controls liver damage in nonalcoholic steatohepatitis. Science 367: 652–660, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Zhao P, Wong KI, Sun X, Reilly SM, Uhm M, Liao Z, Skorobogatko Y, and Saltiel AR. TBK1 at the crossroads of inflammation and energy homeostasis in adipose tissue. Cell 172: 731–743 e12, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Zhao Y, Wang Z, Feng D, Zhao H, Lin M, Hu Y, Zhang N, Lv L, Gao Z, Zhai X, Tian X, and Yao J. p66Shc contributes to liver fibrosis through the regulation of mitochondrial reactive oxygen species. Theranostics 9: 1510–1522, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Zhong X, Huang M, Kim HG, Zhang Y, Chowdhury K, Cai W, Saxena R, Schwabe RF, Liangpunsakul S, and Dong XC. SIRT6 protects against liver fibrosis by deacetylation and suppression of SMAD3 in hepatic stellate cells. Cell Mol Gastroenterol Hepatol 10: 341–364, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]














