Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 49(3); 2025 > Article
Review
Basic and Translational Research Metabolic Sparks in the Liver: Metabolic and Epigenetic Reprogramming in Hepatic Stellate Cells Activation and Its Implications for Human Metabolic Diseases
Yeon Jin Roh*orcid, Hyeonki Kim*orcid, Dong Wook Choiorcidcorresp_icon
Diabetes & Metabolism Journal 2025;49(3):368-385.
DOI: https://doi.org/10.4093/dmj.2025.0195
Published online: May 1, 2025
  • 280 Views
  • 36 Download

Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea

corresp_icon Corresponding author: Dong Wook Choi orcid Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul 02841, Korea E-mail: dongwook_choi@korea.ac.kr
*Yeon Jin Roh and Hyeonki Kim contributed equally to this study as first authors.
• Received: May 9, 2025   • Accepted: April 17, 2025

Copyright © 2025 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

prev next
  • The liver plays a fundamental role in metabolic homeostasis, integrating systemic fuel utilization with the progression of various metabolic diseases. Hepatic stellate cells (HSCs) are a key nonparenchymal cell type in the liver, which is essential for maintaining hepatic architecture in their quiescent state. However, upon chronic liver injury or metabolic stress, HSCs become activated, leading to excessive extracellular matrix deposition and pro-fibrotic signaling, ultimately positioning them as key players in liver pathology. Emerging evidence highlights the critical roles of metabolic reprogramming and epigenetic regulation in HSCs activation. HSCs activation is driven by both intrinsic fuel metabolism reprogramming and extrinsic metabolic cues from the microenvironment, while the metabolic intermediates actively reshape the epigenetic landscape, reinforcing fibrogenic transcriptional programs. In this review, we summarize recent advances in understanding how metabolic and epigenetic alterations drive HSCs activation, thereby shaping transcriptional programs that sustain fibrosis, and discuss potential therapeutic strategies to target these interconnected pathways in human metabolic diseases.
• HSC activation involves coordinated metabolic and epigenetic reprogramming.
• Altered fuel metabolism induces major metabolic shifts in HSCs.
• These shifts alter the epigenetic landscape via histone, DNA, and transcription changes.
• Metabolic-epigenetic changes trigger HSC activation and fibrosis-linked diseases.
The liver plays central roles in systemic homeostasis by regulating fuel utilization, detoxification, bile production, and immune function. Its functional relevance in these processes closely links it to various endocrine and metabolic diseases [1], including metabolic dysfunction-associated fatty liver disease (MAFLD), diabetes, obesity, chronic kidney disease, and cardiovascular diseases.
Among them, MAFLD stands out as a key player, serving as a critical link between metabolic disorders and liver pathology. This condition displays a progressive spectrum, where liver dysfunction and metabolic imbalances fuel disease advancement as MAFLD progresses, liver fibrosis, characterized by excessive extracellular matrix (ECM) accumulation and impaired regenerative capacity, marks as a decisive and irreversible turning point, ultimately leading to significant hepatic dysfunction, and lasting functional impairment. Given its central roles in disease progression, unraveling the mechanisms underlying the pathogenesis of liver fibrosis and developing precise and targeted therapeutic strategies remain essential.
Hepatic stellate cells (HSCs) are key mediators of liver fibrosis. Under normal condition, HSCs resides in the space of Disse, surrounding hepatocytes, and primarily exists in as quiescent HSCs (qHSCs), which play a systemic role in the storage and metabolic regulation of lipids and vitamin A. qHSCs are also essential for maintaining liver homeostasis by interacting with other hepatic cells through various cytokines, chemokines, and growth factors. For instance, qHSCs-derived hepatocyte growth factor modulates Kupffer cell activity by attenuating inflammatory responses, and promotes liver tissue repair [2]. In addition, qHSCs regulate ECM turnover by secreting matrix metalloproteinases which degrade excess ECM and counteract fibrogenic signaling, thereby preventing ECM accumulation and maintaining the liver microenvironment [3].
In response to liver injury, HSCs undergo activation (aHSCs), transitioning from a quiescent to a pro-fibrotic phenotype, a process essential for tissue repair and characterized by increased proliferation, lipid droplet (LD) depletion and enhanced ECM production. However, under condition of chronic liver damage, inflammation, or metabolic imbalance, aHSCs transdifferentiate into myofibroblastic HSCs (mHSCs), which exhibit abberant ECM production and enhanced pro-fibrotic activity, ultimately leading to fibrosis. This phenotypic transition is tightly regulated by a complex network of cytokines, growth factors, and metabolic adaptations, which collectively drive fibrogenic activity.
Recently, the roles of metabolic reprogramming in HSCs activation have gained considerable attention, revealing its impact beyond its traditional bioenergetic function. In this context, alteration of fuel metabolism—such as glycolytic adaptation, glutaminolysis, lipid metabolism, and mitochondrial remodeling—are being actively uncovered [4,5]. Moreoever, emerging evidence underscores the intricate interplay between metabolic changes and epigenetic modifications, where metabolic changes actively remodel epigenetic landscapes, reinforcing fibrogenic gene expression through a positive feedback loop. This metabolism-epigenetics crosstalk has emerged as a key regulatory axis in HSCs activation, influencing signaling pathways beyond direct metabolic adaptation.
In this paper, we aim to provide a comprehensive overview of metabolic alteration occurred during HSCs activation, the relevant epigenetic reprogramming driven by these metabolic changes. Furthermore, we will discuss potential therapeutic strategies for targeting these processes, with a particular focus on precise and targeted approaches to disrupt the fibrogenic cascade, thereby mitigating progression of various metabolic diseases and its complications.
Metabolic reprogramming is widely recognized as a critical driver of HSCs activation and consequent liver fibrosis progression. Accumulating evidence has revealed the pivotal role of altered fuel metabolism in orchestrating the phenotypic transition of qHSCs to aHSCs. This section discusses a thorough overview of the metabolic reprogramming observed in essential fuel metabolism during HSC activation (Fig. 1).
Glucose metabolism
Glucose metabolism serves a fundamental fuel source for virtually all cell types and organisms on earth, integrating various anabolic and catabolic pathways to maintain cellular homeostasis. As a central metabolic hub, glycolysis breaks down glucose to produce adenosine triphosphate (ATP), and provide essential intermediates for biosynthetic processes, including nucleotide, amino acid, and lipid synthesis. Under homeostatic condition, qHSCs primarily rely on oxidative phosphorylation to maintain energy homeostasis. However, upon activation, they undergo a metabolic switch to aerobic glycolysis, a shift that provides both rapid energy production and biosynthetic support for proliferation and ECM production. This metabolic adaption characterized by increased glucose uptake and its conversion to lactate, even in the presence of oxygen is a hallmark of the Warburg effect, a phenomenon primarily observed in cancer cells.
The glycolytic reprogramming of HSCs is driven by coordinated transcriptional and metabolic events. Primarily, transcriptional activation of glucose transporters (GLUTs), including GLUT1, GLUT2, and GLUT4 significantly increase glucose uptake [6,7]. Expression of key rate limiting enzymes, such as hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2), is also significantly upregulated, contributing to the enhanced glycolytic flux [8]. In contrast, gluconeogenic enzymes including phosphoenolpyruvate carboxykinase (PCK1) and fructose bisphosphatase (FBP1) are downregulated, thereby reinforcing the glycolytic phenotype.
Reprogramming of glucose metabolism also include various signaling pathways. For example, Hedgehog (Hh) signaling through Gli and leptin converge upon the transcription factor hypoxia-inducible factor 1α (HIF-1α), a master regulator of metabolic changes, upregulating expression of GLUTs and glycolytic enzymes.
Additionally, platelet-derived growth factor (PDGF) upregulates the phosphoinositide 3-kinase (PI3K)-protein kinase B (Akt)-mechanistic target of rapamycin (mTOR) signaling cascade, thereby enhancing the expression of key glycolytic enzymes, including GLUT1 and PKM2. Notably, PDGF activation of glycolytic flux is further amplified by the production of Extracellular vesicles (EV) containing glycolysis-related enzymes, such as GLUT1 and PKM2, which transfers to other qHSCs. These transferred EVs ultimately modulate the glucose metabolism and promoting activation [9], thereby contributing to the heterogeneity of HSCs activation between the pericentral and periportal regions.
Lactate, the main product of glycolysis catalyzed by lactate dehydrogenase (LDH), may also play a significant role in HSCs activation. In aHSCs, the high glycolytic flux leads to lactate accumulation, and the lactate stabilizes HIF-1α by inhibiting prolyl hydroxylases, creating a feed-forward loop that sustains glycolytic flux [10]. Additionally, lactate can be exported via monocarboxylate transporter 4 (MCT4), establishing an acidic niche that promotes pro-fibrotic M2 macrophage polarization by inducing α-ketoglutarate (α-KG) production [10].
Glutamine metabolism
Glutamine is an amino acid that serves as a critical metabolic substrate in various cellular processes, including energy production, nucleotide biosynthesis, and redox homeostasis. In rapidly proliferating cells, glutamine metabolism plays key roles in sustaining anabolic and bioenergetic demands.
In aHSCs, glutaminolysis, the conversion of glutamine to α-KG, is significantly elevated [11]. The accumulation of α-KG as a product of the increased glutaminolysis, supports proliferation and fibrogenic activity by serving as a fuel for the tricarboxylic acid (TCA) cycle, particularly when glucose is predominantly utilized for aerobic glycolysis.
Glutamine uptake in HSCs is primarily mediated by the solute carrier family 1 member 5 (SLC1A5) transporter, whose expression increases during activation [12]. Once inside the cell, glutamine is converted to glutamate, catalyzed by glutaminase (GLS), the rate limiting step of glutaminolysis. Glutamate is then further processed by glutamate dehydrogenase (GLUD1) or transaminases (GOT1/GOT2) and converted to α-KG, which enters the TCA cycle. aHSCs exhibit upregulated expressions of GLS, GLUD1, and GOT1/2 at both mRNA and protein levels, correlating with increased α-KG flux. Several signaling pathways act as upstream regulators of the glutaminolysis in this context. For example, Yes-associated protein 1 (YAP)/WW-domain-containing transcription regulator 1 (TAZ), which mediates cell proliferation and apoptosis, promotes the transcription of glutaminolytic enzymes, including GLS and GOT1, by binding to transcriptional enhanced associated domain (TEAD) family proteins [13,14]. Hh signaling cooperates with YAP to amplify glutaminolytic flux. When the Hh pathway is activated, its downstream smoothened (SMO) facilitates the expression and nuclear localization of YAP [14,15]. This activation promotes the transcription of YAP target genes, thereby reinforcing the glutaminolytic phenotype of activated HSCs. Blocking Hh-YAP axis has been shown to prevent liver fibrosis; genetic ablation of YAP or pharmacological inhibition (e.g., verteporfin) reduces glutamine uptake and α-KG levels, impairing HSCs proliferation [16]. In murine models, Hh inhibition (e.g., cyclopamine) or HSCs specific SMO knockout attenuates GLS activity and fibrosis progression [15,17,18]. However, some studies have reported contradictory findings where dimethyl α-ketoglutarate, a cell-permeable ester form of α-KG, displays inhibitory effects on fibrosis and autophagy in HSCs [19,20]. One possible explanation is that extracellular α-KG plays distinct roles compared to intracellular α-KG.
Glutamine can also serve as a precursor for proline synthesis, contributing to the production of collagen, a major component of the ECM in liver fibrosis [21,22]. The increased glutamine conversion to proline supports the fibrogenic activity of HSCs. Additionally, the downregulation of proline oxidase (POX) expression during HSCs activation may lead to an accumulation of free proline, further reinforcing collagen synthesis and deposition [23].
Lipid and vitamin A metabolism
qHSCs account for approximately 80% to 90% of vitamin A storage and regulation in the body, serving as the primary systemic reservoir of vitamin A [24]. Vitamin A is not merely stored in HSCs, it actively participate in maintaining HSCs quiescence [25-27].
Vitamin A, in the form of retinoids conjugated to retinol-binding protein, is transported into HSCs through stimulated by retinoic acid gene 6. Once internalized, lecithin retinol acyltransferase (LRAT) esterifies retinoids with fatty acid to form retinyl esters (RE), which are subsequently stored in LDs [28]. In addition to RE, LDs contain triglycerides and cholesterol esters serving as energy reservoirs [29,30]. These droplets are highly dynamic organelles, with their composition and turnover closely linked to HSCs activation. Upon activation, HSCs lose their LDs, consequently their stored vitamin A, leading to enhanced fibrogenic activity; LDs are mobilized and transported to autophagosomes, where they undergo lipophagy, a selective form of autophagy that degrades LDs to release free fatty acids (FFAs) and free cholesterol [31-34]. The protein autophagy-related gene 7 (ATG7) plays a crucial role in this process by facilitating the formation of autophagosomes [35]. Once released, the FFAs are transported to mitochondria, where they undergo β-oxidation to generate ATP, providing the energy required for HSCs proliferation and ECM production. In this context, β-oxidation, a metabolic pathway that integrates long chain fatty acid (LCFA) into mitochondrial metabolism, is upregulated in aHSCs reinforcing the metabolic shift from lipid storage to energy production, thereby supporting fibrosis as an energy-intensive processes. Additionally, retinyl ester hydrolase (REH) promotes the breakdown of fatty acids bound to REs, regenerating retinol. Retinol is further catabolized into retinoic acid (RA), a bioactive metabolite of vitamin A that acts as a signaling molecule to maintain the quiescent phenotype of HSCs [36,37]. Retinoic acid receptors (RAR) and retinoid X receptors (RXR) are nuclear receptors activated by retinoids and play significant roles in suppressing fibrogenic genes [38,39].
Elevated free cholesterol levels contribute to HSCs activation by enhancing the expression of toll-like receptor 4 (TLR4), a pattern recognition receptor that increases the cells sensitivity to transforming growth factor-beta (TGF-β), a key cytokine that drives fibrogenesis [40]. This heightened sensitivity amplifies HSCs responses to fibrogenic stimuli, leading to ECM deposition and tissue remodeling. Cholesterol accumulation in HSCs is also regulated by low-density lipoprotein receptor (LDLR), which facilitates cholesterol uptake of cholesterol from circulating low-density lipoproteins (LDL), and miR-33a, a microRNA that modulates cholesterol homeostasis by suppressing cholesterol efflux [41]. Furthermore, oxidized LDL (oxLDL), a modified form of LDL, has been shown to induce HSCs activation through Wnt signaling pathway, further reinforcing fibrogenesis [42]. OxLDL also binds to scavenger receptors on the HSCs surface, triggering intracellular signaling cascades that upregulate fibrogenic gene expression and enhance collagen production [43]. Targeting cholesterol metabolism in HSCs may represent a promising therapeutic approach for targeting liver fibrosis [44]. This is further supported by interesting observations showing that inhibition of acyl-CoA: cholesterol acyltransferase 1 (ACAT1), an enzyme responsible for the esterification of free cholesterol, mitigates free cholesterol accumulation in HSCs, thereby suppressing fibrotic progression [45].
Two major transcription factors are known to regulate lipid metabolism in qHSCs; peroxisome proliferator-activated receptor gamma (PPAR-γ) and sterol regulatory element-binding protein 1 (SREBP1) [46,47]. PPAR-γ is activated with its specific ligands such as fatty acids, eicosanoids and functions as heterodimer with RXR [38]. This heterodimer complex translocates to the nucleus and upregulates fat storage gene expression by binding to the peroxisome proliferator-responsive element sequence. SREBP is a master regulator of fatty acid and lipid synthesis. Under normal condition, SREBP remains in an inactive state in the endoplasmic reticulum (ER) membrane, bound to SREBP cleavage-activating protein (SCAP) and insulin-induced gene (INSIG), which acts as lipid sensors. However, when intracellular levels of fatty acids and cholesterol decline, ubiquitin-X domain-containing protein 8 (UBXD8) recognizes INSIG and promotes its ubiquitination and extraction from the ER membrane, leading to its proteasomal degradation [48]. As a result, SREBP is liberated from the complex, translocates to the golgi where it undergoes proteolytic cleavage by site-1, 2 proteases to generate its functional form. This cleaved transcription factor, corresponding to the N-terminal portion of SREBP, binds to sterol regulatory element (SRE), facilitating transcription of lipid synthesis genes.
During HSCs activation, the expression of PPAR-γ decreases [49], whereas SREBP is upregulated in aHSCs and facilitates lipid remodeling that supports ER expansion and cell proliferation. In particular, SREBP2 plays a key role in maintaining cholesterol homeostasis essential for preserving the quiescent phenotype of HSCs [50,51], Disruptions in this pathway result in the accumulation of free cholesterol, thereby promoting HSC activation [52,53]. Notably, several studies have shown that reintroducing PPAR-γ into aHSCs can restore them to a quiescent, less fibrogenic state, indicating its potential as a therapeutic target [54,55]. Furthermore, pharmacological or genetic inhibition of SREBP signaling has been shown to effectively suppress HSC activation [56,57]. Collectively, these findings suggest that targeting lipid metabolism through the modulation of key transcription factors such as PPAR-γ and SREBP holds therapeutic promise for the treatment of liver fibrosis.
As previously stated, HSCs activation is accompanied by substantial metabolic alterations. Emerging evidence emphasizes the intricate crosstalk between metabolic alterations and epigenetic modifications as the key drivers of phenotypic switch of quiescent, vitamin A-storing cells to proliferative, contractile, ECM-producing aHSCs. This crosstalk collectively regulates the transcriptional reprogramming necessary for HSCs activation.
In this section, we specifically focus on how metabolic alterations shape the epigenetic landscape during HSCs activation, through various post-translational modifications of histones and DNA, as well as the regulation of nuclear receptors (Fig. 2).
Histone acetylation
Histone modifications play pivotal roles in regulating chromatin structure and gene expression. Various metabolic substrates are used to modify the histone tails and the structured globular domain of nucleosome core particles. Histone acetylation, in particular, is a critical epigenetic modification mediated by a wide range of histone acetyltransferases (HATs) This process alters chromatin architecture by relaxing its structure, allowing for transcription factors access to DNA, thereby promoting gene expression.
Enhanced glycolysis in activated HSCs serves as a predominant fuel source for the synthesis of acetyl-coenzyme A (CoA), a metabolic substrate of histone acetylation [58]. A recent study has shown that PDGF-stimulated glycolysis in aHSCs fuels histone acetylation, particularly at histone H3 lysine 9 (H3K9), on the promoter regions of EV-related genes, leading to increased expression of genes, such as ras related protein 31 [9]. This epigenetic changes result in the increased release of EVs, which are enriched with fibrogenic proteins and amplify liver fibrosis [9]. Conversely, sirtuins (SIRTs), which exerts deacetylase activity in an nicotinamide adenine dinucleotide, oxidized form-dependent manner, also link cellular metabolism to epigenetic states. Given the possibility that increased glycolytic flux in aHSCs leads to NAD+ depletion, aHSCs may exhibit impaired SIRT activity, thereby contributing to fibrotic phenotypes through enhanced histone acetylation [59].
These functional connection between glucose metabolism and histone acetylation is further elaborated upon by a line of investigation demonstrating that specific glycolytic enzymes, such as PKM2 and LDH modulates the metabolic-epigenetic link, and inhibition or modulation of these enzymes attenuate HSCs activation and liver fibrosis [60,61].
For example, dimerized PKM2, which is significantly enriched in aHSCs, promotes aerobic glycolysis, and upregulates specific transcription factors via histone H3 modification, thus driving HSCs activation [60]. However, PKM2 tetramerization, induced by the allosteric agent thienopyrrolopyridazinone, inhibits HSCs activation by reducing aerobic glycolysis and decreasing expression of myelocytomatosis oncogene and cyclin D1 through the regulation of histone H3K9 acetylation. Both TEPP-46 and the PKM2 inhibitor attenuated liver fibrosis in vivo in a mouse model [60].
Similarly, LDH inhibition protects against hepatic fibrosis by regulating the metabolic reprogramming of HSCs in both in vitro and in vivo models [61]. Its protective effects are presumably mediated through disrupted NAD+ recycle and reduced nuclear lactate levels, both of which are linked to potential epigenetic changes. In particular, since lactate is an endogenous inhibitor of histone deacetylase (HDAC), LDH inhibition also increases HDAC activity, followed by decrease of histone acetylation, particularly at the c-Myc promoter region [61]. The downregulation of c-Myc further suppresses the expression of GLS2 and the glutamine transporter (SLC1A5), thereby restricting glutaminolysis and contributing to HSCs inactivation [61].
These findings suggest that metabolic reprogramming during HSCs activation induces histone acetylation, which in turn drive additional metabolic changes, creating a feedback loop that amplifies HSCs activation and fibrosis progression.
Histone lactylation
Histone lactylation is another histone modification that links metabolic reprogramming to HSCs activation. Traditionally considered a by-product of glycolysis, lactate has recently been recognized as a direct regulator of gene transcription by modifying histone lysine residues at target gene promoter regions. A recent study demonstrated that insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) functions a key regulator in HSCs activation and liver fibrosis by upregulating aldolase A, a key glycolytic enzyme, thereby enhancing glycolytic flux into lactate production [62]. Furthermore, MCTs, especially MCT1, facilitate the intracellular and potentially nuclear transport of lactate, enabling lactate accumulation in the nucleus [63]. Once inside the nucleus, lactate can be locally converted to lactyl-CoA by nuclear-localized acyl-CoA synthetases or metabolized by nuclear LDH activity, providing substrates for histone lactylation [64,65]. This leads to elevated histone lactylation in aHSCs, particularly H3K18 lactylation (H3K18la), which is enriched in the promoter regions of genes associated with HSCs activation. Inhibition of glycolysis using 2-deoxy-D-glucose reduces histone lactylation in vitro and ameliorates liver fibrosis by suppressing HSCs activation in vivo [62].
Similarly, HK2 expression also increases during HSCs activation, contributing to lactate accumulation and histone H3K18la [66]. Deletion of the Hk2 gene or inhibition of lactate production reduces H3K18la and suppresses HSCs activation, while, exogenous lactate, but not acetate, rescues the activating capacities of Hk2-deficient HSCs. Notably, class 1 HDAC inhibition increases H3K18ac, whereas reducing H3K18la, ultimately suppressing HSCs activation [66]. These findings revealed the competitive relationship between histone lactylation and acetylation in the regulation of HSCs activation. However, as previously mentioned, histone acetylation has also been implicated in promoting HSCs activation; Therefore, the intricate balance between lactylation and acetylation may play a crucial role in determining HSCs activation and fibrosis progression.
Beyond histone lactylation, various metabolic byproducts can also link metabolic reprogramming to epigenetic modifications [67]. For instance, β-hydroxybutyrylation (BHB), a systemic fuel source under starvation condition, fuels histone BHBylation, which is associated with gene activation in response to energy stress and metabolic adaptation [68]. Given that HSCs undergo significant metabolic shifts during activation and are located adjacent to hepatocytes, the primary site of BHB production, BHB may influence fibrogenic gene expression and cellular plasticity by manipulating epigenetic landscapes. Further studies are needed to elucidate the role of metabolite-driven histone modifications and their balance in HSCs activation and liver fibrosis progression.
DNA methylation
HSCs activation is accompanied by extensive remodeling of the DNA methylation landscape, driven by increased DNA methyltransferase (DNMT) activity and reduced expression of ten-eleven translocation (TET) enzyme which functions as a DNA demethylase [69]. This shift leads to elevated promoter methylation of anti-fibrotic genes, suppressing their expression, while simultaneously reducing methylation of pro-fibrotic genes, leading to their upregulation. Concurrently, global levels of DNA hydroxy-methylation decline, limiting the dynamic regulation of DNA methylation [69]. Genome-wide analyses have revealed distinct DNA methylation patterns between qHSCs and aHSCs, suggesting that epigenetic regulation of DNA methylation plays a crucial role in HSCs activation and fibrosis progression [69]. HSCs activation entails a metabolic rewiring of the methionine cycle that influences the cellular S-adenosylmethionine (SAM)/S-adenosylhomocysteine (SAH) ratio, also known as the methylation potential. Hepatocytes predominantly express methionine adenosyltransferase 1A (MAT1A), which encodes methionine adenosyltransferase (MAT) enzymes responsible for SAM synthesis. However, during hepatic fibrosis and HSCs activation, MAT1A expression decreases whereas MAT2A and MAT2B, which encode alternative MAT isoforms, are upregulated [70]. MAT2A encodes the catalytic β2 subunit of MATII, the enzyme responsible for SAM biosynthesis in HSCs, while MAT2B encodes a regulatory α subunit, which binds to MAT2A to form a heterooligomeric complex. This interaction increases feedback inhibition by SAM, leading to reduced MATII enzymatic activity and lower intracellular SAM levels, despite the upregulation of MATII subunits [70]. The decline in SAM levels reduces the SAM/SAH ratio, influencing gene expression and promoting a fibrogenic phenotype. This paradox—higher MAT2A/B expression but lower SAM availability—is a hallmark of chronic liver fibrosis [70]. Silencing MAT2A in activated HSCs further depletes SAM and suppresses proliferation, indicating that a sufficient SAM pool is necessary for maintaining the activated phenotype [70]. Conversely, perturbations in methionine/SAM metabolism alter DNA methylation, where a higher SAM/SAH ratio favors methylation and a more qHSCs state, while SAM depletion or SAH accumulation inhibits DNMT activity, leading to hypomethylation of fibrogenic genes [70]. A recent study showed that oroxylin A, a flavonoid derived from Scutellaria baicalensis with anti-fibrotic properties, inhibits MAT2A expression, reducing SAM levels, and suppressing DNMT3A activity, leading to cyclic GMP-AMP synthase (cGAS) gene hypomethylation [71]. As a result, cGAS-stimulator of interferon genes (STING) signaling is activated, promoting HSCs senescence, which serves as a novel anti-fibrotic mechanism [71].
During activation, HSCs generate α-KG through glutaminolysis, which is essential for maintaining the activity of α-KG–dependent dioxygenases, including TET enzymes which catalyze DNA demethylation by oxidizing 5-methylcytosine (5mC) [72]. Sufficient α-KG facilitates the demethylation of promoter regions, whereas α-KG depletion or accumulation of succinate and 2-hydroxyglutarate, the metabolic byproducts that inhibit α-KG–dependent dioxygenases activity, can impair TET activity, leading to aberrant DNA hypermethylation [73]. However, direct evidence demonstrating that α-KG drives the demethylation specifically at pro-fibrotic gene promoters in HSCs remains limited. While α-KG is known to support TET enzyme activity broadly, further research is required to confirm whether it directly modulates the epigenetic regulation of fibrogenic genes in HSCs. Nonetheless, given its role in DNA demethylation, α-KG may contribute to the activation of fibrogenic genes in HSCs. Thus, glutamine metabolism, which primarily regulates the α-KG pool in HSCs, may influence the epigenetic landscape and activation of fibrogenic pathways in HSCs.
Nuclear receptor mediated epigenetic modifications
As previously mentioned, HSCs activation is characterized by the loss of LDs, leading to the subsequent release of retinoids and FFAs [74]. RA, derived from RE, binds to RARs (RARα/β/γ), which dimerize with RXR to regulate gene transcription [75].
In qHSCs, ligand-unbound RAR/RXR complexes recruit corepressors nuclear receptor corepressor (NcoR)/silencer mediator of retinoid and thyroid hormone receptor (SMRT), HDACs and polycomb repressive complexes (PRC2), leading to chromatin condensation and gene silencing via histone modifications such as H3K27 trimethylation (H3K27me3) [76-79]. This repressive state maintains quiescence-associated genes, including PPARγ and RARβ. However, as RA level increase during HSCs activation, the complex undergoes a conformational change that releases corepressors and recruits coactivators such as p300/CREB-binding protein, which possess HAT activity [80]. This shift promotes histone acetylation (H3/H4ac) and the increase of gene-activating marks such as H3K4 tri-methylation (H3K4me3), opening chromatin structure and allowing transcription of RA-target genes that counteract early-stage HSCs activation. The subsequent loss of RA during later stage HSCs activation diminishes this transcriptional control, leading to hypermethylation of quiescence-associated genes including PPARγ and RARβ, reinforcing the fibrogenic phenotype [81].
Additionally, FFAs serve as ligands for nuclear receptors, including PPARs and RXR [75]. In qHSCs, PPARγ forms a heterodimer with RXR to regulate lipid metabolism and storage, thereby suppressing HSCs activation. However, during HSCs activation, PPARγ expression is epigenetically repressed through DNA hypermethylation and histone modifications such as H3K9me3, H3K27me3, at its promoter regions, leading to the loss of FFA-mediated anti-fibrotic signaling [79]. Notably, certain polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA) and arachidonic acid (AA), can directly bind and activate RXR, regulating gene expression. RXR, as a heterodimeric partner for multiple nuclear receptors, including PPAR, farnesoid X receptor, liver X receptor, RAR, functions as a central hub for shaping lipid metabolism and fibrosis regulation [82]. The alteration of FFA availability during HSCs activation modulates HSCs activation through disruption of RXR signaling, followed by epigenetic changes.
As discussed in the previous sections, metabolic reprogramming in HSCs drives their transition into aHSCs, by (1) fueling biosynthetic pathways and mitochondrial energetics, and (2) shaping the epigenetic landscape, both of which are closely linked to signaling pathways and transcriptional regulation. Given these tightly interconnected mechanisms, pharmacological interventions targeting both metabolic reprogramming and fibrogenic signaling in HSCs may represent promising therapeutic strategies. This section explores how different pharmacological agents inhibit HSCs activation, primarily from a metabolic perspective and within disease-specific contexts, with a focus on their mechanistic actions in type 2 diabetes mellitus (T2DM) and other components of metabolic syndrome including obesity, adipose tissue dysfunction, and hypertension (Table 1).
Type 2 diabetes mellitus
T2DM is a complex metabolic disorder characterized by multiple symptoms including chronic hyperglycemia, insulin resistance, and adipokine imbalances, which collectively drive HSCs activation; High blood glucose levels result in oxidative stress and formation of advanced glycation end-products (AGEs), which binds to receptor for AGEs (RAGE) expressed on HSCs, triggering fibrotic responses [83,84].
Insulin resistance in T2DM leads to excess FFAs and hepatic steatosis, causing lipotoxic hepatocyte injury and release of pro-fibrotic cytokines such as TGF-β1 [85,86]. In this environment, HSCs undergo activation, and subsequently produce collagen, a key structural component of the ECM. Adipokine imbalance may further contribute to HSCs activation. A study has shown that obese T2DM patients often have elevated leptin (a pro-fibrogenic adipokine) and reduced adiponectin (an anti-fibrotic adipokine), presumably shifting HSCs toward an activated state [87], as leptin directly stimulates HSCs via Janus tyrosine kinase (JAK)/signal transducer and activator of transcription (STAT) and TGF-β pathways, whereas adiponectin normally induces PPARγ in HSCs to maintain their quiescence [88-90]. Collectively, the metabolic disturbance creates a persistently pro-fibrotic milieu in T2DM, thereby driving HSCs activation, and fibrosis progression.
Several antidiabetic agents mitigate pro-fibrotic pathways in T2DM by directly targeting HSCs activation. For example, Metformin has been shown to activate the energy sensor AMP-activated protein kinase (AMPK) in HSCs, which in turn counteract HSCs activation by reducing alpha-smooth muscle actin (α-SMA) expression and inhibiting HSCs proliferation [91]. Mechanistically, AMPK activation by metformin disrupts the succinate–succinate-G protein-coupled receptor 91 (GPR91) signaling axis, a metabolic pathway that drives HSCs activation, thereby attenuating mHSCs trans-differentiation [92]. In addition, metformin reduces AGE accumulation and RAGE signaling, leading to the suppression of nuclear factor kappa B (NF-κB)-mediated inflammatory signaling in HSCs. Through these mechanisms, metformin alleviates hyperglycemia-driven oxidative stress and AGE/RAGE pathways, both of which contribute to HSCs activation in T2DM.
Thiazolidinediones (TZDs), such as rosiglitazone and pioglitazone, are PPARγ agonists that directly target HSCs’ transcriptional program [93]. In T2DM, aHSCs exerts a reduced PPARγ expression and activity, whereas TZD treatment reactivates PPARγ, inducing an adipogenic gene profile that mitigates fibrogenesis. Additionally, in a rat HSCs cell line, rosiglitazone treatment upregulates PPARγ expression and significantly decreases α-SMA and type I collagen synthesis [94]. Moreover, it also inhibits HSCs proliferation and even promotes apoptosis of activated HSCs. Interestingly, this mechanism overlaps with adiponectin’s anti-fibrotic effect, as adiponectin requires intact PPARγ signaling to exert its suppressive effects on HSCs activation [95]. Thus, TZDs pharmacologically mimic the adiponectin-PPARγ axis to reverse HSCs activation in T2DM.
Sodium-glucose cotransporter 2 (SGLT2) inhibitors (e.g., empagliflozin) have been reported to exert liver-protective, anti-fibrotic effects beyond glucose control. Specifically, studies have shown that SGLT2 inhibition attenuates HSCs activation primarily through metabolic and signaling pathways.
For example, empagliflozin has been shown to downregulate miR-34a-5p, a microRNA highly enriched in HSCs from a mouse model of diabetic liver fibrosis that promotes TGF-β signaling [96]. By lowering miR-34a, empagliflozin restores the expression of its target gene gremlin 2, an antagonist of TGF-β, thereby reducing Smad-mediated fibrogenic gene expression [96].
SGLT2 inhibitors have also been shown to interfere with the Hh-YAP pathway in HSCs [97], In addition, empagliflozin treatment in fibrotic rats suppressed key components of Hh signaling pathway, including Patched1, SMO, Gli2, thereby mitigating this pro-fibrotic metabolic programming [98]. By inhibiting Hh-YAP signals, SGLT2 inhibitors reduce HSCs glutaminolysis and proliferation, contributing to fibrosis attenuation [98]. This observation aligns with the previously discussed intricate connection between Hh-YAP axis and glutamine metabolism. Taken together, SGLT2 inhibitors directly target HSCs activation in T2DM, by inhibiting miR-34a-TGF-β and Hh-YAP axis, exerting metabolic and transcriptional modulation.
Dipeptidyl peptidase-4 inhibitors (DPP-4i), such as sitagliptin, gemigliptin and vildagliptin, have also been shown to exhibit anti-fibrotic activity in diabetes-related liver disease by targeting HSCs activation [99-101]. Studies have demonstrated that sitagliptin suppresses HSCs activation by inhibiting multiple kinase cascade pathways [99]. In aHSCs, DPP-4 inhibition attenuated PDGF-BB-induced proliferation, and reduced TGF-1 signaling and collagen I gene expression. Mechanistically, sitagliptin prevented the phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase and of Smad2/3, the key effectors of TGF-β signaling pathway [99]. Inhibiting p38 and Smad2/3 activation interrupts the pro-fibrotic gene program in aHSCs and attenuates the trans-differentiation. DPP-4is also mitigate the insulin resistance-associated inflammation and TGF-β signaling pathway, both of which drive HSCs activation. Notably, DPP-4i also increase endogenous glucagon-like peptide-1 (GLP-1) levels, which may further benefit the liver by reducing lipotoxicity and inflammation.
GLP-1 receptor agonists (e.g., liraglutide) have emerged as anti-fibrotic agents due to their effects on hepatic metabolism and inflammation. While GLP-1 agonists are primarily known for improving insulin sensitivity and promoting weight loss, they have also been shown to directly modulate stress pathways in HSCs [102].
In a study using diet-induced nonalcoholic steatohepatitis (NASH) models, liraglutide lowered reactive oxygen species (ROS) levels and suppressed the upregulation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1/2/4 (NOX1/2/4) NADPH oxidases and RAGE in the liver, thereby attenuating HSCs activation. Liraglutide’s anti-fibrotic effect was also confirmed in vitro, where it was blunted upon RAGE or NOX2 overexpression, indicating that the mechanisms involves disrupting the ROS–RAGE feed-forward loop that drives HSCs activation [102]. GLP-1 agonism also shifts macrophages toward an anti-inflammatory phenotype and may enhance adiponectin release, further alleviating HSCs activation [103]. Through these mechanisms including reducing ROS, inhibiting RAGE/NF-κB signaling, and improving the metabolic environments, GLP-1 receptor agonists disrupt key links between T2DM and HSCs activation.
Other components of metabolic syndrome
Metabolic syndrome encompasses obesity, insulin resistance, adipose dysfunction, and hypertension which play key roles in creating a pathological milieu that drives HSCs activation [104,105]. These metabolic disturbances coordinately converge to generate lipotoxic, inflammatory, and hormonal signals, ultimately, contributing to liver fibrosis. While the precise molecular mechanisms remain under investigation, several key biological components, including metabolism and the relevant signaling pathways, are implicated in these processes, offering potential targets for therapeutic intervention in HSCs activation and liver fibrosis.
Obesity and adipose tissue dysfunction
Excess caloric intake leads to dysregulated hepatic lipid metabolism, which drives HSCs activation through oxidative stress and inflammation, representing a pivotal mechanisms in the pathogenesis of liver fibrosis [105]. In addition to these inflammatory signals, chronic overnutrition induces systemic insulin resistance and lipid overload, leading to shifts in hepatic fuel metabolism, such as enhanced glycolysis, glutaminolysis, and lipid accumulation [9,106,107]. These metabolic alterations not only provide biosynthetic precursors and energy that fuel fibrogenic processes but also influence epigenetic regulatory mechanisms, thereby sustaining HSCs activation.
The hepatic accumulation of FFAs and cholesterol results in peroxidation injury, releasing damage-associated molecular patterns (DAMPs) that activate Kupffer cells and recruit inflammatory cells [108,109]. This process is exacerbated by lipid peroxidation products and ROS, which stimulate TLR4 on both Kupffer cells and HSCs, triggering NF-κB-mediated inflammatory signaling [40,110].
Additionally, adipose tissue dysfunction plays a critical role in HSCs activation. Visceral fat produces excess pro-inflammatory adipokines (e.g., leptin, resistin) while only few adipokines with anti-inflammatory properties (e.g., adiponectin) [111,112]. For example, high leptin levels derived from visceral fat in obesity have pro-fibrogenic effects by directly activating HSCs and increasing TGF-β1 secretion, thereby promoting collagen production [111]. Conversely, hypoadiponectinemia removes an important brake on HSCs activation, since adiponectin normally upregulates PPARγ in stellate cells to maintain their quiescent, lipid-storing state [112].
Statins (3-hydroxy-3-methylglutaryl coenzyme A [HMG-CoA] reductase inhibitors), while developed for dyslipidemia, also exhibit anti-fibrotic properties in the liver [113]. One mechanism involves induction of ferroptosis, an iron-dependent form of cell death triggered by lipid peroxidation. aHSCs are susceptible to ferroptosis when their antioxidant defenses are impaired. Statins, by blocking the mevalonate pathway, deplete downstream products, such as geranylgeranyl pyrophosphate and coenzyme Q10, which are crucial for neutralizing lipid ROS in HSCs [113]. Simvastatin, a commonly used lipophilic statin, enhances lipid peroxidation of HSCs, thereby inducing ferroptotic cell death. Consequently, statin treatment makes HSCs more sensitive to ferroptosis and ameliorates experimental fibrosis [113]. Additionally, statins’ pleiotropic effects improve endothelial function and reduce hepatic inflammation, indirectly inactivating HSCs. There is also evidence that statins may modulate PPAR signaling in the liver [114]. For instance, some class of statins can activate PPARα, improving fatty acid oxidation and thereby reducing lipotoxic stress on the liver. Additionally, statins’ pleiotropic effects improve endothelial function and reduce hepatic inflammation, indirectly inactivating HSCs. Taken together, the lipid-lowering, anti-oxidative, and HSC-cytotoxic effects of statins help counteract the lipid-driven HSC activation in metabolic syndrome.
In the context of aberrant inflammation, novel anti-inflammatory agents targeting HSC-intrinsic stress pathways are under investigation. One example is J2H-1702, a potent 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitor [115]. 11β-HSD1 in liver tissue generates active cortisol, which can drive gluconeogenesis and also local inflammation. In HSCs, excess glucocorticoid signaling via 11β-HSD1 may support a pro-fibrogenic state in aHSCs by interacting with inflammatory pathways. In a recent study, inhibiting 11β-HSD1 with J2H-1702 ‘reprogrammed’ HSCs, decreasing their activation markers and inflammatory signaling [115]. The mechanism underlying J2H-1702 inhibition of HSCs activation also involves suppressed phosphorylated Smad3 (downstream of TGF-β) and reduced NF-κB-related gene expression, which led to lower α-SMA and collagen I expression, indicating HSCs were reverting toward quiescence. In diet-induced NASH models, J2H-1702 significantly alleviates liver steatosis and fibrosis, supporting its therapeutic potential. Given the 11β-HSD1 role in promoting hepatic gluconeogenesis, J2H-1702 may indirectly exert its suppressive effects on glycolytic flux in HSCs, thereby reinforcing HSCs inhibition. This exemplifies a strategy of targeting intra-hepatic inflammation and metabolism to prevent HSC activation.
Therapeutic interventions in metabolic syndrome aim to target these upstream drivers to mitigate HSCs activation. Strategies to amplify adiponectin signaling or PPARγ activity can reverse the HSC activation due to obesity and adipose tissue dysfunction. Adiponectin itself has been shown to protect against fibrosis; transgenic mice overexpressing adiponectin are resistant to liver fibrosis, whereas adiponectin-knockout mice develop more severe fibrosis [116]. Adiponectin binding to its receptors on HSCs activates AMP kinase and stimulates PPARγ expression, which reprograms HSCs toward a less fibrogenic, more lipid-storing phenotype [116]. Pharmacologically, this is mimicked by PPARγ agonists (TZDs) and newer selective PPARγ modulators. For example, the experimental modulator SR1664 significantly reduced fibrosis in obese mouse models without typical TZD side effects [117]. SR1664 and similar agents directly act on HSCs to induce PPARγ-target genes, thereby inhibiting collagen deposition [117]. In diet-induced NASH, SR1664 decreased activated HSCs numbers (α-SMA-positive cells) and collagen I content, confirming that PPARγ reactivation can therapeutically disable HSCs [117]. Thus, pharmacological restoration of the adiponectin-PPARγ axis, either directly or indirectly, may address the adipokine deficiency that drives HSCs activation in metabolic syndrome.
Hypertension
The renin-angiotensin system (RAS) is often overactive in metabolic syndrome primarily due to adipose-derived angiotensinogen and systemic hypertension, leading to elevated angiotensin II (Ang II) [118]. Elevated Ang II levels in hypertension stimulate glycolysis and mitochondrial ROS production in HSCs via angiotensin II type 1 (AT1) receptors, fueling fibrogenic processes via TGF-β1/Smad3 signaling [119]. Additionally, systemic insulin resistance associated with hypertension leads to lactate accumulation, which induces histone lactylation and sustains HSCs activation [120]. These findings suggest that hypertension is not only a cardiovascular disorder but also a metabolic driver of liver fibrosis, emphasizing the need for therapeutic strategies targeting hypertension-induced metabolic and epigenetic dysregulation in HSCs.
Renin-angiotensin system blockers (ARBs) attenuate fibrosis by disrupting Ang II/AT1 pro-fibrotic signaling in HSCs [121]. Losartan, an AT1 receptor antagonist, has demonstrated anti-fibrotic efficacy in both animal models and clinical studies. By blocking AT1 receptor on HSCs, losartan prevents Ang II-induced NOX activation, calcium influx, and TGF-β1 upregulation, thereby inhibiting HSCs activation [121]. In a T cell-mediated fibrosis model, losartan treatment alleviates fibrogenesis and significantly downregulated TGF-β1 expression, resulting in reduced HSCs activation and proliferation [121]. Losartan also appears to exert PPARγ-agonistic effects, further suppressing HSC activation [122].
Similarly, perindopril, an angiotensin-converting enzyme (ACE) inhibitor, achieves upstream blockade of Ang II production. In bile duct-ligated rats, perindopril markedly reduced hepatic hydroxyproline content and collagen gene expression, whereas reducing number of HSCs that produce α-SMA [123]. Perindopril also diminished oxidative stress markers in the liver [123].
Taken together, by relieving HSCs from continuous Ang II stimulation, ARBs and ACE inhibitor reduce the contractile and pro-fibrotic phenotype of HSCs by disrupting the Ang II–TGF-β loop of HSCs activation, offering therapeutic benefits for patients with metabolic syndrome.
Liver fibrosis, driven by HSC activation, represents a critical and irreversible stage in the progression of a myriad of metabolic diseases. These metabolic diseases are commonly associated with chronic metabolic dysregulation including high blood glucose level, insulin resistance, lipid accumulation, and systemic inflammation, all of which significantly contribute to HSCs activation. Once activated, HSCs further exacerbate these metabolic disturbances, thereby creating a pathological cycle that accelerates disease progression. This review highlights recent findings on the central role of metabolic reprogramming in HSCs activation, where alterations in various aspects of fuel utilization, including glycolysis, glutaminolysis, and lipid metabolism, contribute to the pro-fibrotic phenotype. Furthermore, the interplay between metabolism and epigenetic modifications further reinforces the fibrogenic cascade, establishing a feed-forward loop that sustains disease progression.
While various omics approaches including transcriptomics, epigenetics, and proteomics have been extensively applied to elucidate the mechanisms underlying HSC activation, metabolomics-based investigations remain largely unexplored. Given the metabolic plasticity of HSCs, precise metabolomic approaches are essential to fully understand the metabolic drivers of HSC activation. Stable isotope tracing analysis, both in vivo and in vitro, particularly in primary HSCs and mouse models of various metabolic diseases, may be essential for precisely mapping fuel metabolism dynamics and identify key metabolic flux alterations in aHSCs. Additionally, nuclear metabolomics can provide critical insights into how metabolic changes influence epigenetic regulation, which is largely determined by the availability of nuclear metabolic substrates for histone and DNA modification. This approach provides a deeper understanding of the tightly interconnected epigenetic and transcriptional landscape that mediates HSCs activation. However, due to the inherently low abundance of HSCs in the liver, traditional bulk metabolomics approaches may lack the sensitivity required for accurately capturing cell-type-specific metabolic rewiring. Therefore, the adoption of advanced single-cell metabolomics techniques will be required for dissecting the heterogeneity of HSCs metabolic states in both in vivo and in vitro settings. Future research should focus on integrating these advanced metabolomics approaches with other omics platforms to provide a comprehensive understanding of HSCs activation, ultimately facilitating the development of targeted and effective therapeutic strategies for liver fibrosis and metabolic diseases.

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

FUNDING

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (2022R1C1C1007023 and 2022M3A9B6017654) and the Ministry of Education (RS-2024-00415102).

Acknowledgements
None
Fig. 1.
Metabolic changes and fuel utilization in hepatic stellate cells (HSCs) activation. Quiescent HSCs play a crucial role in storing lipids and vitamins through the regulation of peroxisome proliferator-activated receptor gamma (PPARγ) and sterol regulatory element-binding protein (SREBP). During the activation into activation HSC, various signals, such as transforming growth factor-beta (TGF-β) and platelet-derived growth factor (PDGF), trigger metabolic changes to provide the energy and metabolic intermediates required for fibrogenesis. Key characteristics of this process include enhanced aerobic glycolysis, glutaminolysis, and lipolysis. Each signal is marked with a dot on the arrows where it induces metabolic changes. HGF, hepatocyte growth factor; MMP, matrix metalloproteinase; STRA6, stimulated by retinoic acid gene 6; REH, retinyl ester hydrolase; LRAT, lecithin retinol acyltransferase; SCAP, SREBP cleavage-activating protein; INSIG, insulin-induced gene; ER, endoplasmic reticulum; RE, retinyl esters; RXR, retinoid X receptor; MCT, monocarboxylate transporter; GLUT, glucose transporter; TCA, tricarboxylic acid; α-KG, α-ketoglutarate; RARβ, retinoic acid receptor β.
dmj-2025-0195f1.jpg
Fig. 2.
Metabolic reprogramming-driven epigenetic modifications in hepatic stellate cells (HSCs) activation. Intricate crosstalk between metabolic reprogramming and epigenetic modifications is essential in HSCs activation. Key metabolic alterations, including enhanced glycolysis, glutaminolysis, and loss of lipid droplets, drive histone and DNA modifications that regulate HSCs activation and fibrosis progression. Histone acetylation, lactylation, and DNA methylation are dictated by availability of metabolic intermediates such as acetyl-coenzyme A (CoA), lactate, S-adenosylmethionine (SAM), and α-ketoglutarate (α-KG). Additionally, nuclear receptor signaling modulates chromatin remodeling in response to retinoic acid and free fatty acids, thereby reinforcing transcriptional programs associated with HSCs activation. NAD+, nicotinamide adenine dinucleotide; NADH, nicotinamide adenine dinucleotide, reduced form; LDH, lactate dehydrogenase; HK2, hexokinase 2; PKM2, pyruvate M2; SIRT, sirtuin; HDAC, histone deacetylase; HAT, histone acetyltransferases; TCA, tricarboxylic acid; TET, ten-eleven translocation; CBP, CREB-binding protein; RAR, retinoic acid receptor; RXR, retinoid X receptor; NCoR, nuclear receptor corepressor; SMRT, silencer mediator of retinoid and thyroid hormone receptor; RE, retinyl esters; DNMT, DNA methyltransferase; MAT, methionine adenosyltransferase; SAH, S-adenosylhomocysteine.
dmj-2025-0195f2.jpg
dmj-2025-0195f3.jpg
Table 1.
Pharmacological strategies for hepatic stellate cell activation in metabolic diseases
Causes of HSCs activation Therapeutics
Reference
Medications Mechanisms
T2DM
 Hyperglycemia Metformin AMPK↑, mTOR↓, succinate-GPR91↓, electron transport chain↓ [85,86]
 Insulin resistance Thiazolidinediones (Pioglitazone) PPAR-γ↑ [87-89]
 Adipokines dysregulation SGLT2 inhibitor (Empagliflozin, Tofogliflozin) miR-34a-5p↓, hedgehog-YAP pathway↓, Hippo signaling↑ [90-92]
 AGE/RAGE signaling DPP-4 inhibitors ERK1/2, p38 and Smad2/3 phosphorylation↓, mitochondrial dysfunction↓, ROS↓ [93-95]
GLP-1 receptor agonists RAGE/NOX2↓, ROS↓ [96,97]
Obesity
 α-SMA/MMP9/STAT3 signaling Statin Modulating PPAR balance, Ferroptosis↑ [107,108]
 Inflamation J2H-1702 NF-κB signaling↓, Smad3/TGF-β signaling↓ [109]
 TGF-β signaling
 Lipotoxicity Adiponectin, SR1664 PPAR-γ↑ [110,111]
Hypertension
 Oxidative stress Losartan NADPH oxidase↓, ROS↓ [115,116]
 Renin-angiotensin system Perindopril Inhibiting ACE [117]

HSC, hepatic stellate cell; T2DM, type 2 diabetes mellitus; AGE/RAGE, advanced glycation end-products/receptor for advanced glycation endproducts; AMPK, AMP-activated protein kinase; mTOR, mechanistic target of rapamycin; GPR91, succinate-G protein-coupled receptor 91; PPAR-γ, peroxisome proliferator-activated receptor gamma; SGLT2, sodium-glucose cotransporter 2; YAP, Yes-associated protein; DPP-4, dipeptidyl peptidase-4; ERK1/2, extracellular signal-regulated kinase 1/2; ROS, reactive oxygen species; GLP-1, glucagon-like peptide-1; NOX2, NADPH oxidase 2; α-SMA, alpha-smooth muscle actin; MMP9, matrix metalloproteinase-9; STAT3, signal transducer and activator of transcription 3; TGF-β, transforming growth factor-beta; NF-κB, nuclear factor kappa B; NADPH, nicotinamide adenine dinucleotide phosphate; ACE, angiotensin-converting enzyme.

  • 1. Alamri ZZ. The role of liver in metabolism: an updated review with physiological emphasis. Int J Basic Clin Pharmacol 2018;7:2271-6.ArticlePDF
  • 2. Kamm DR, McCommis KS. Hepatic stellate cells in physiology and pathology. J Physiol 2022;600:1825-37.ArticlePubMedPDF
  • 3. Benyon RC, Arthur MJ. Extracellular matrix degradation and the role of hepatic stellate cells. Semin Liver Dis 2001;21:373-84.ArticlePubMed
  • 4. Trivedi P, Wang S, Friedman SL. The power of plasticity-metabolic regulation of hepatic stellate cells. Cell Metab 2021;33:242-57.ArticlePubMed
  • 5. Petersen MC, Vatner DF, Shulman GI. Regulation of hepatic glucose metabolism in health and disease. Nat Rev Endocrinol 2017;13:572-87.ArticlePubMedPMCPDF
  • 6. Chandrashekaran V, Das S, Seth RK, Dattaroy D, Alhasson F, Michelotti G, et al. Purinergic receptor X7 mediates leptin induced GLUT4 function in stellate cells in nonalcoholic steatohepatitis. Biochim Biophys Acta 2016;1862:32-45.ArticlePubMed
  • 7. Lin J, Chen A. Curcumin diminishes the impacts of hyperglycemia on the activation of hepatic stellate cells by suppressing membrane translocation and gene expression of glucose transporter-2. Mol Cell Endocrinol 2011;333:160-71.ArticlePubMed
  • 8. Mejias M, Gallego J, Naranjo-Suarez S, Ramirez M, Pell N, Manzano A, et al. CPEB4 increases expression of PFKFB3 to induce glycolysis and activate mouse and human hepatic stellate cells, promoting liver fibrosis. Gastroenterology 2020;159:273-88.ArticlePubMed
  • 9. Khanal S, Liu Y, Bamidele AO, Wixom AQ, Washington AM, Jalan-Sakrikar N, et al. Glycolysis in hepatic stellate cells coordinates fibrogenic extracellular vesicle release spatially to amplify liver fibrosis. Sci Adv 2024;10:eadn5228.ArticlePubMedPMC
  • 10. Kozlov AM, Lone A, Betts DH, Cumming RC. Lactate preconditioning promotes a HIF-1α-mediated metabolic shift from OXPHOS to glycolysis in normal human diploid fibroblasts. Sci Rep 2020;10:8388.ArticlePubMedPMCPDF
  • 11. Du K, Chitneni SK, Suzuki A, Wang Y, Henao R, Hyun J, et al. Increased glutaminolysis marks active scarring in nonalcoholic steatohepatitis progression. Cell Mol Gastroenterol Hepatol 2020;10:1-21.ArticlePubMed
  • 12. Scalise M, Pochini L, Console L, Losso MA, Indiveri C. The human SLC1A5 (ASCT2) amino acid transporter: from function to structure and role in cell biology. Front Cell Dev Biol 2018;6:96.ArticlePubMedPMC
  • 13. Yu HX, Yao Y, Bu FT, Chen Y, Wu YT, Yang Y, et al. Blockade of YAP alleviates hepatic fibrosis through accelerating apoptosis and reversion of activated hepatic stellate cells. Mol Immunol 2019;107:29-40.ArticlePubMed
  • 14. Du K, Hyun J, Premont RT, Choi SS, Michelotti GA, Swiderska-Syn M, et al. Hedgehog-YAP signaling pathway regulates glutaminolysis to control activation of hepatic stellate cells. Gastroenterology 2018;154:1465-79.e13.ArticlePubMed
  • 15. Chen Y, Choi SS, Michelotti GA, Chan IS, Swiderska-Syn M, Karaca GF, et al. Hedgehog controls hepatic stellate cell fate by regulating metabolism. Gastroenterology 2012;143:1319-29.e11.ArticlePubMed
  • 16. Konishi T, Schuster RM, Lentsch AB. Proliferation of hepatic stellate cells, mediated by YAP and TAZ, contributes to liver repair and regeneration after liver ischemia-reperfusion injury. Am J Physiol Gastrointest Liver Physiol 2018;314:G471-82.ArticlePubMedPMC
  • 17. Simon J, Nunez-Garcia M, Fernandez-Tussy P, Barbier-Torres L, Fernandez-Ramos D, Gomez-Santos B, et al. Targeting hepatic glutaminase 1 ameliorates non-alcoholic steatohepatitis by restoring very-low-density lipoprotein triglyceride assembly. Cell Metab 2020;31:605-22.e10.ArticlePubMedPMC
  • 18. Feng J, Wang C, Liu T, Li J, Wu L, Yu Q, et al. Procyanidin B2 inhibits the activation of hepatic stellate cells and angiogenesis via the Hedgehog pathway during liver fibrosis. J Cell Mol Med 2019;23:6479-93.ArticlePubMedPMCPDF
  • 19. Zhao J, Peng L, Luo Z, Cui R, Yan M. Inhibitory effects of dimethyl α-ketoglutarate in hepatic stellate cell activation. Int J Clin Exp Pathol 2015;8:5471-7.PubMedPMC
  • 20. Zhao J, Peng L, Cui R, Guo X, Yan M. Dimethyl α-ketoglutarate reduces CCl4-induced liver fibrosis through inhibition of autophagy in hepatic stellate cells. Biochem Biophys Res Commun 2016;481:90-6.ArticlePubMed
  • 21. Karna E, Miltyk W, Wolczynski S, Palka JA. The potential mechanism for glutamine-induced collagen biosynthesis in cultured human skin fibroblasts. Comp Biochem Physiol B Biochem Mol Biol 2001;130:23-32.ArticlePubMed
  • 22. Krane SM. The importance of proline residues in the structure, stability and susceptibility to proteolytic degradation of collagens. Amino Acids 2008;35:703-10.ArticlePubMedPDF
  • 23. Li J, Ghazwani M, Liu K, Huang Y, Chang N, Fan J, et al. Regulation of hepatic stellate cell proliferation and activation by glutamine metabolism. PLoS One 2017;12:e0182679.ArticlePubMedPMC
  • 24. Yoneda A, Sakai-Sawada K, Niitsu Y, Tamura Y. Vitamin A and insulin are required for the maintenance of hepatic stellate cell quiescence. Exp Cell Res 2016;341:8-17.ArticlePubMed
  • 25. Amengual J, Zhang N, Kemerer M, Maeda T, Palczewski K, Von Lintig J. STRA6 is critical for cellular vitamin A uptake and homeostasis. Hum Mol Genet 2014;23:5402-17.ArticlePubMedPMC
  • 26. Blaner WS, Hendriks HF, Brouwer A, de Leeuw AM, Knook DL, Goodman DS. Retinoids, retinoid-binding proteins, and retinyl palmitate hydrolase distributions in different types of rat liver cells. J Lipid Res 1985;26:1241-51.ArticlePubMed
  • 27. Kawaguchi R, Yu J, Honda J, Hu J, Whitelegge J, Ping P, et al. A membrane receptor for retinol binding protein mediates cellular uptake of vitamin A. Science 2007;315:820-5.ArticlePubMed
  • 28. Blaner WS, van Bennekum AM, Brouwer A, Hendriks HF. Distribution of lecithin-retinol acyltransferase activity in different types of rat liver cells and subcellular fractions. FEBS Lett 1990;274:89-92.ArticlePubMedPDF
  • 29. Yamada M, Blaner WS, Soprano DR, Dixon JL, Kjeldbye HM, Goodman DS. Biochemical characteristics of isolated rat liver stellate cells. Hepatology 1987;7:1224-9.ArticlePubMed
  • 30. Moriwaki H, Blaner WS, Piantedosi R, Goodman DS. Effects of dietary retinoid and triglyceride on the lipid composition of rat liver stellate cells and stellate cell lipid droplets. J Lipid Res 1988;29:1523-34.ArticlePubMed
  • 31. Zhang Z, Zhao S, Yao Z, Wang L, Shao J, Chen A, et al. Autophagy regulates turnover of lipid droplets via ROS-dependent Rab25 activation in hepatic stellate cell. Redox Biol 2017;11:322-34.PubMed
  • 32. Thoen LF, Guimaraes EL, Dolle L, Mannaerts I, Najimi M, Sokal E, et al. A role for autophagy during hepatic stellate cell activation. J Hepatol 2011;55:1353-60.ArticlePubMed
  • 33. Hernandez-Gea V, Ghiassi-Nejad Z, Rozenfeld R, Gordon R, Fiel MI, Yue Z, et al. Autophagy releases lipid that promotes fibrogenesis by activated hepatic stellate cells in mice and in human tissues. Gastroenterology 2012;142:938-46.ArticlePubMed
  • 34. Singh R, Kaushik S, Wang Y, Xiang Y, Novak I, Komatsu M, et al. Autophagy regulates lipid metabolism. Nature 2009;458:1131-5.ArticlePubMedPMCPDF
  • 35. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell 2019;176:11-42.ArticlePubMedPMC
  • 36. Shajari S, Saeed A, Smith-Cortinez NF, Heegsma J, Sydor S, Faber KN. Hormone-sensitive lipase is a retinyl ester hydrolase in human and rat quiescent hepatic stellate cells. Biochim Biophys Acta Mol Cell Biol Lipids 2019;1864:1258-67.ArticlePubMed
  • 37. Schreiber R, Taschler U, Preiss-Landl K, Wongsiriroj N, Zimmermann R, Lass A. Retinyl ester hydrolases and their roles in vitamin A homeostasis. Biochim Biophys Acta 2012;1821:113-23.ArticlePubMedPMC
  • 38. Berry DC, Noy N. All-trans-retinoic acid represses obesity and insulin resistance by activating both peroxisome proliferation-activated receptor beta/delta and retinoic acid receptor. Mol Cell Biol 2009;29:3286-96.PubMedPMC
  • 39. Berry DC, DeSantis D, Soltanian H, Croniger CM, Noy N. Retinoic acid upregulates preadipocyte genes to block adipogenesis and suppress diet-induced obesity. Diabetes 2012;61:1112-21.ArticlePubMedPMCPDF
  • 40. Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA, et al. TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med 2007;13:1324-32.ArticlePubMedPDF
  • 41. Li ZJ, Ou-Yang PH, Han XP. Profibrotic effect of miR-33a with Akt activation in hepatic stellate cells. Cell Signal 2014;26:141-8.ArticlePubMed
  • 42. Myung SJ, Yoon JH, Gwak GY, Kim W, Lee JH, Kim KM, et al. Wnt signaling enhances the activation and survival of human hepatic stellate cells. FEBS Lett 2007;581:2954-8.ArticlePubMed
  • 43. Schneiderhan W, Schmid-Kotsas A, Zhao J, Grunert A, Nussler A, Weidenbach H, et al. Oxidized low-density lipoproteins bind to the scavenger receptor, CD36, of hepatic stellate cells and stimulate extracellular matrix synthesis. Hepatology 2001;34(4 Pt 1):729-37.ArticlePubMed
  • 44. Kang Q, Chen A. Curcumin eliminates oxidized LDL roles in activating hepatic stellate cells by suppressing gene expression of lectin-like oxidized LDL receptor-1. Lab Invest 2009;89:1275-90.ArticlePubMedPMCPDF
  • 45. Tomita K, Teratani T, Suzuki T, Shimizu M, Sato H, Narimatsu K, et al. Acyl-CoA:cholesterol acyltransferase 1 mediates liver fibrosis by regulating freecholesterol accumulation in hepatic stellate cells. J Hepatol 2014;61:98-106.ArticlePubMed
  • 46. She H, Xiong S, Hazra S, Tsukamoto H. Adipogenic transcriptional regulation of hepatic stellate cells. J Biol Chem 2005;280:4959-67.ArticlePubMed
  • 47. Tsukamoto H. Adipogenic phenotype of hepatic stellate cells. Alcohol Clin Exp Res 2005;29(11 Suppl):132S-3S.ArticlePubMed
  • 48. Lee JN, Zhang X, Feramisco JD, Gong Y, Ye J. Unsaturated fatty acids inhibit proteasomal degradation of Insig-1 at a postubiquitination step. J Biol Chem 2008;283:33772-83.ArticlePubMedPMC
  • 49. Yan K, Deng X, Zhai X, Zhou M, Jia X, Luo L, et al. p38 mitogen-activated protein kinase and liver X receptor-α mediate the leptin effect on sterol regulatory element binding protein1c expression in hepatic stellate cells. Mol Med 2012;18:10-8.ArticlePubMedPDF
  • 50. Bengoechea-Alonso MT, Ericsson J. SREBP in signal transduction: cholesterol metabolism and beyond. Curr Opin Cell Biol 2007;19:215-22.ArticlePubMed
  • 51. Radhakrishnan A, Goldstein JL, McDonald JG, Brown MS. Switch-like control of SREBP-2 transport triggered by small changes in ER cholesterol: a delicate balance. Cell Metab 2008;8:512-21.ArticlePubMedPMC
  • 52. Tomita K, Teratani T, Suzuki T, Shimizu M, Sato H, Narimatsu K, et al. Free cholesterol accumulation in hepatic stellate cells: mechanism of liver fibrosis aggravation in nonalcoholic steatohepatitis in mice. Hepatology 2014;59:154-69.ArticlePubMed
  • 53. Teratani T, Tomita K, Suzuki T, Oshikawa T, Yokoyama H, Shimamura K, et al. A high-cholesterol diet exacerbates liver fibrosis in mice via accumulation of free cholesterol in hepatic stellate cells. Gastroenterology 2012;142:152-64.e10.ArticlePubMed
  • 54. Marra F, Efsen E, Romanelli RG, Caligiuri A, Pastacaldi S, Batignani G, et al. Ligands of peroxisome proliferator-activated receptor gamma modulate profibrogenic and proinflammatory actions in hepatic stellate cells. Gastroenterology 2000;119:466-78.ArticlePubMed
  • 55. Hazra S, Xiong S, Wang J, Rippe RA, Krishna V, Chatterjee K, et al. Peroxisome proliferator-activated receptor gamma induces a phenotypic switch from activated to quiescent hepatic stellate cells. J Biol Chem 2004;279:11392-401.PubMed
  • 56. Lee EH, Lee JH, Kim DY, Lee YS, Jo Y, Dao T, et al. Loss of SREBP-1c ameliorates iron-induced liver fibrosis by decreasing lipocalin-2. Exp Mol Med 2024;56:1001-12.ArticlePubMedPMCPDF
  • 57. Lemoine M, Barbu V, Girard PM, Kim M, Bastard JP, Wendum D, et al. Altered hepatic expression of SREBP-1 and PPARgamma is associated with liver injury in insulin-resistant lipodystrophic HIV-infected patients. AIDS 2006;20:387-95.PubMed
  • 58. Egger AS, Rauch E, Sharma S, Kipura T, Hotze M, Mair T, et al. Linking metabolism and histone acetylation dynamics by integrated metabolic flux analysis of Acetyl-CoA and histone acetylation sites. Mol Metab 2024;90:102032.ArticlePubMedPMC
  • 59. Guo X, Kesimer M, Tolun G, Zheng X, Xu Q, Lu J, et al. The NAD(+)-dependent protein deacetylase activity of SIRT1 is regulated by its oligomeric status. Sci Rep 2012;2:640.ArticlePubMedPMCPDF
  • 60. Zheng D, Jiang Y, Qu C, Yuan H, Hu K, He L, et al. Pyruvate kinase M2 tetramerization protects against hepatic stellate cell activation and liver fibrosis. Am J Pathol 2020;190:2267-81.ArticlePubMedPMC
  • 61. Li L, Lei Q, Zhen Y, Cao L, Dong Y, Liu X, et al. Lactate dehydrogenase inhibition protects against hepatic fibrosis by regulating metabolic reprogramming of hepatic stellate cells. J Agric Food Chem 2024;72:27953-64.ArticlePubMedPDF
  • 62. Zhou Y, Yan J, Huang H, Liu L, Ren L, Hu J, et al. The m6A reader IGF2BP2 regulates glycolytic metabolism and mediates histone lactylation to enhance hepatic stellate cell activation and liver fibrosis. Cell Death Dis 2024;15:189.ArticlePubMedPMCPDF
  • 63. Min K, Yenilmez B, Kelly M, Echeverria D, Elleby M, Lifshitz LM, et al. Lactate transporter MCT1 in hepatic stellate cells promotes fibrotic collagen expression in nonalcoholic steatohepatitis. Elife 2024;12:RP89136.PubMedPMC
  • 64. Zhu R, Ye X, Lu X, Xiao L, Yuan M, Zhao H, et al. ACSS2 acts as a lactyl-CoA synthetase and couples KAT2A to function as a lactyltransferase for histone lactylation and tumor immune evasion. Cell Metab 2025;37:361-76.e7.ArticlePubMed
  • 65. Liu R, Ren X, Park YE, Feng H, Sheng X, Song X, et al. Nuclear GTPSCS functions as a lactyl-CoA synthetase to promote histone lactylation and gliomagenesis. Cell Metab 2025;37:377-94.e9.ArticlePubMed
  • 66. Rho H, Terry AR, Chronis C, Hay N. Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis. Cell Metab 2023;35:1406-23.e8.ArticlePubMedPMC
  • 67. Dai Z, Ramesh V, Locasale JW. The evolving metabolic landscape of chromatin biology and epigenetics. Nat Rev Genet 2020;21:737-53.ArticlePubMedPMCPDF
  • 68. Zhou T, Cheng X, He Y, Xie Y, Xu F, Xu Y, et al. Function and mechanism of histone β-hydroxybutyrylation in health and disease. Front Immunol 2022;13:981285.ArticlePubMedPMC
  • 69. Page A, Paoli P, Moran Salvador E, White S, French J, Mann J. Hepatic stellate cell transdifferentiation involves genome-wide remodeling of the DNA methylation landscape. J Hepatol 2016;64:661-73.ArticlePubMed
  • 70. Ramani K, Yang H, Kuhlenkamp J, Tomasi L, Tsukamoto H, Mato JM, et al. Changes in the expression of methionine adenosyltransferase genes and S-adenosylmethionine homeostasis during hepatic stellate cell activation. Hepatology 2010;51:986-95.ArticlePubMed
  • 71. Zhao D, Gao Y, Su Y, Zhou Y, Yang T, Li Y, et al. Oroxylin A regulates cGAS DNA hypermethylation induced by methionine metabolism to promote HSC senescence. Pharmacol Res 2023;187:106590.ArticlePubMed
  • 72. Tran KA, Dillingham CM, Sridharan R. The role of α-ketoglutarate-dependent proteins in pluripotency acquisition and maintenance. J Biol Chem 2019;294:5408-19.ArticlePubMed
  • 73. Cui W, Huang Z, Pfeifer GP. Lack of major genome-wide DNA methylation changes in succinate-treated human epithelial cells. Int J Mol Sci 2022;23:5663.ArticlePubMedPMC
  • 74. Tuohetahuntila M, Molenaar MR, Spee B, Brouwers JF, Wubbolts R, Houweling M, et al. Lysosome-mediated degradation of a distinct pool of lipid droplets during hepatic stellate cell activation. J Biol Chem 2017;292:12436-48.ArticlePubMedPMC
  • 75. Sharvit E, Abramovitch S, Reif S, Bruck R. Amplified inhibition of stellate cell activation pathways by PPAR-γ, RAR and RXR agonists. PLoS One 2013;8:e76541.ArticlePubMedPMC
  • 76. Loinder K, Soderstrom M. The nuclear receptor corepressor (N-CoR) modulates basal and activated transcription of genes controlled by retinoic acid. J Steroid Biochem Mol Biol 2003;84:15-21.ArticlePubMed
  • 77. Urvalek AM, Gudas LJ. Retinoic acid and histone deacetylases regulate epigenetic changes in embryonic stem cells. J Biol Chem 2014;289:19519-30.ArticlePubMedPMC
  • 78. Gudas LJ, Wagner JA. Retinoids regulate stem cell differentiation. J Cell Physiol 2011;226:322-30.ArticlePubMedPMC
  • 79. Jiang Y, Xiang C, Zhong F, Zhang Y, Wang L, Zhao Y, et al. Histone H3K27 methyltransferase EZH2 and demethylase JMJD3 regulate hepatic stellate cells activation and liver fibrosis. Theranostics 2021;11:361-78.ArticlePubMedPMC
  • 80. Dietze EC, Caldwell LE, Marcom K, Collins SJ, Yee L, Swisshelm K, et al. Retinoids and retinoic acid receptors regulate growth arrest and apoptosis in human mammary epithelial cells and modulate expression of CBP/p300. Microsc Res Tech 2002;59:23-40.ArticlePubMed
  • 81. Kweon SM, Chi F, Higashiyama R, Lai K, Tsukamoto H. Wnt pathway stabilizes MeCP2 protein to repress PPAR-γ in activation of hepatic stellate cells. PLoS One 2016;11:e0156111.ArticlePubMedPMC
  • 82. Lengqvist J, Mata De Urquiza A, Bergman AC, Willson TM, Sjovall J, Perlmann T, et al. Polyunsaturated fatty acids including docosahexaenoic and arachidonic acid bind to the retinoid X receptor alpha ligand-binding domain. Mol Cell Proteomics 2004;3:692-703.PubMed
  • 83. Kiss K, Regos E, Rada K, Firneisz G, Baghy K, Kovalszky I. Chronic hyperglycaemia induced alterations of hepatic stellate cells differ from the effect of TGFB1, and point toward metabolic stress. Pathol Oncol Res 2020;26:291-9.ArticlePubMedPDF
  • 84. Xia JR, Chen TT, Li WD, Lu FL, Liu J, Cai XG, et al. Inhibitory effect of receptor for advanced glycation end product-specific small interfering RNAs on the development of hepatic fibrosis in primary rat hepatic stellate cells. Mol Med Rep 2015;12:569-74.ArticlePubMed
  • 85. Dong Z, Zhuang Q, Ning M, Wu S, Lu L, Wan X. Palmitic acid stimulates NLRP3 inflammasome activation through TLR4-NF-κB signal pathway in hepatic stellate cells. Ann Transl Med 2020;8:168.ArticlePMC
  • 86. Gharbia S, Nazarie SR, Dinescu S, Balta C, Herman H, Peteu VE, et al. Adipose-derived stem cells (ADSCs) supplemented with hepatocyte growth factor (HGF) attenuate hepatic stellate cell activation and liver fibrosis by inhibiting the TGF-β/ Smad signaling pathway in chemical-induced liver fibrosis associated with diabetes. Cells 2022;11:3338.ArticlePubMedPMC
  • 87. Lee MW, Lee M, Oh KJ. Adipose tissue-derived signatures for obesity and type 2 diabetes: adipokines, batokines and microRNAs. J Clin Med 2019;8:854.ArticlePubMedPMC
  • 88. Gu YJ, Sun WY, Zhang S, Li XR, Wei W. Targeted blockade of JAK/STAT3 signaling inhibits proliferation, migration and collagen production as well as inducing the apoptosis of hepatic stellate cells. Int J Mol Med 2016;38:903-11.ArticlePubMed
  • 89. Elinav E, Ali M, Bruck R, Brazowski E, Phillips A, Shapira Y, et al. Competitive inhibition of leptin signaling results in amelioration of liver fibrosis through modulation of stellate cell function. Hepatology 2009;49:278-86.ArticlePubMed
  • 90. Beltagy DM, GamalAbdelsalam K, Mohamed TM, El-Keey MM. Role of harmaline as adiponectin modulator in defeating liver cirrhosis induced by thioacetamide in mice. Biomed Pharmacol J 2021;14:123-31.Article
  • 91. Li Z, Ding Q, Ling LP, Wu Y, Meng DX, Li X, et al. Metformin attenuates motility, contraction, and fibrogenic response of hepatic stellate cells in vivo and in vitro by activating AMP-activated protein kinase. World J Gastroenterol 2018;24:819-32.ArticlePubMedPMC
  • 92. Nguyen G, Park SY, Le CT, Park WS, Choi DH, Cho EH. Metformin ameliorates activation of hepatic stellate cells and hepatic fibrosis by succinate and GPR91 inhibition. Biochem Biophys Res Commun 2018;495:2649-56.ArticlePubMed
  • 93. Galli A, Crabb DW, Ceni E, Salzano R, Mello T, Svegliati-Baroni G, et al. Antidiabetic thiazolidinediones inhibit collagen synthesis and hepatic stellate cell activation in vivo and in vitro. Gastroenterology 2002;122:1924-40.ArticlePubMed
  • 94. Yang H, Zhao LF, Zhang L, Zhang XH, Zhang XQ. Rosiglitazone inhibits hepatic stellate cell proliferation by regulating peroxisome proliferator-activated receptor gamma/ heme oxygenase-1 expression. Zhonghua Gan Zang Bing Za Zhi 2020;28:410-5.PubMed
  • 95. Shafiei MS, Shetty S, Scherer PE, Rockey DC. Adiponectin regulation of stellate cell activation via PPARγ-dependent and -independent mechanisms. Am J Pathol 2011;178:2690-9.ArticlePubMedPMC
  • 96. Shen Y, Cheng L, Xu M, Wang W, Wan Z, Xiong H, et al. SGLT2 inhibitor empagliflozin downregulates miRNA-34a5p and targets GREM2 to inactivate hepatic stellate cells and ameliorate non-alcoholic fatty liver disease-associated fibrosis. Metabolism 2023;146:155657.ArticlePubMed
  • 97. Abdalla N, Abo-ElMatty DM, Saleh S, Ghattas MH, Omar NN. Empagliflozin suppresses hedgehog pathway, alleviates ER stress, and ameliorates hepatic fibrosis in rats. Sci Rep 2023;13:19046.ArticlePubMedPMCPDF
  • 98. Heo YJ, Lee N, Choi SE, Jeon JY, Han SJ, Kim DJ, et al. Empagliflozin reduces the progression of hepatic fibrosis in a mouse model and inhibits the activation of hepatic stellate cells via the hippo signalling pathway. Biomedicines 2022;10:1032.ArticlePubMedPMC
  • 99. Kaji K, Yoshiji H, Ikenaka Y, Noguchi R, Aihara Y, Douhara A, et al. Dipeptidyl peptidase-4 inhibitor attenuates hepatic fibrosis via suppression of activated hepatic stellate cell in rats. J Gastroenterol 2014;49:481-91.ArticlePubMedPDF
  • 100. Nguyen G, Park SY, Do DV, Choi DH, Cho EH. Gemigliptin alleviates succinate-induced hepatic stellate cell activation by ameliorating mitochondrial dysfunction. Endocrinol Metab (Seoul) 2022;37:918-28.ArticlePubMedPMCPDF
  • 101. Khalil R, Shata A, Abd El-Kader EM, Sharaf H, Abdo WS, Amin NA, et al. Vildagliptin, a DPP-4 inhibitor, attenuates carbon tetrachloride-induced liver fibrosis by targeting ERK1/2, p38α, and NF-κB signaling. Toxicol Appl Pharmacol 2020;407:115246.ArticlePubMed
  • 102. Ji J, Feng M, Huang Y, Niu X. Liraglutide inhibits receptor for advanced glycation end products (RAGE)/reduced form of nicotinamide-adenine dinucleotide phosphate (NAPDH) signaling to ameliorate non-alcoholic fatty liver disease (NAFLD) in vivo and vitro. Bioengineered 2022;13:5091-102.ArticlePubMedPMC
  • 103. Simental-Mendia LE, Sanchez-Garcia A, Linden-Torres E, Simental-Mendia M. Impact of glucagon-like peptide-1 receptor agonists on adiponectin concentrations: a meta-analysis of randomized controlled trials. Br J Clin Pharmacol 2021;87:4140-9.ArticlePubMedPDF
  • 104. Friedman SL. Hepatic fibrosis and cancer: the silent threats of metabolic syndrome. Diabetes Metab J 2024;48:161-9.ArticlePubMedPMCPDF
  • 105. Yu Y, Liu Y, An W, Song J, Zhang Y, Zhao X. STING-mediated inflammation in Kupffer cells contributes to progression of nonalcoholic steatohepatitis. J Clin Invest 2019;129:546-55.ArticlePubMed
  • 106. Yin X, Dong L, Wang X, Qin Z, Ma Y, Ke X, et al. Perilipin 5 regulates hepatic stellate cell activation and high-fat diet-induced non-alcoholic fatty liver disease. Animal Model Exp Med 2024;7:166-78.ArticlePubMed
  • 107. Yin X, Peng J, Gu L, Liu Y, Li X, Wu J, et al. Targeting glutamine metabolism in hepatic stellate cells alleviates liver fibrosis. Cell Death Dis 2022;13:955.ArticlePubMedPMCPDF
  • 108. Wang YH, Twu YC, Wang CK, Lin FZ, Lee CY, Liao YJ. Niemann-Pick type C2 protein regulates free cholesterol accumulation and influences hepatic stellate cell proliferation and mitochondrial respiration function. Int J Mol Sci 2018;19:1678.ArticlePubMedPMC
  • 109. Zelber-Sagi S, Ivancovsky-Wajcman D, Fliss-Isakov N, Hahn M, Webb M, Shibolet O, et al. Serum malondialdehyde is associated with non-alcoholic fatty liver and related liver damage differentially in men and women. Antioxidants (Basel) 2020;9:578.ArticlePubMedPMC
  • 110. Lan T, Kisseleva T, Brenner DA. Deficiency of NOX1 or NOX4 prevents liver inflammation and fibrosis in mice through inhibition of hepatic stellate cell activation. PLoS One 2015;10:e0129743.ArticlePubMedPMC
  • 111. Saxena NK, Ikeda K, Rockey DC, Friedman SL, Anania FA. Leptin in hepatic fibrosis: evidence for increased collagen production in stellate cells and lean littermates of ob/ob mice. Hepatology 2002;35:762-71.ArticlePubMedPDF
  • 112. Li N, Zhao S, Zhang Z, Zhu Y, Gliniak CM, Vishvanath L, et al. Adiponectin preserves metabolic fitness during aging. Elife 2021;10:e65108.ArticlePubMedPMCPDF
  • 113. Kitsugi K, Noritake H, Matsumoto M, Hanaoka T, Umemura M, Yamashita M, et al. Simvastatin inhibits hepatic stellate cells activation by regulating the ferroptosis signaling pathway. Biochim Biophys Acta Mol Basis Dis 2023;1869:166750.ArticlePubMed
  • 114. Feng J, Dai W, Mao Y, Wu L, Li J, Chen K, et al. Simvastatin resensitizes hepatocellular carcinoma cells to sorafenib by inhibiting HIF-1α/PPAR-γ/PKM2-mediated glycolysis. J Exp Clin Cancer Res 2020;39:24.ArticlePubMedPMCPDF
  • 115. Lee SY, Kim S, Choi I, Song Y, Kim N, Ryu HC, et al. Inhibition of 11β-hydroxysteroid dehydrogenase 1 relieves fibrosis through depolarizing of hepatic stellate cell in NASH. Cell Death Dis 2022;13:1011.ArticlePubMedPMCPDF
  • 116. Ramezani-Moghadam M, Wang J, Ho V, Iseli TJ, Alzahrani B, Xu A, et al. Adiponectin reduces hepatic stellate cell migration by promoting tissue inhibitor of metalloproteinase-1 (TIMP1) secretion. J Biol Chem 2015;290:5533-42.ArticlePubMedPMC
  • 117. McVicker BL, Hamel FG, Simpson RL, Bennett RG. A selective PPARγ modulator reduces hepatic fibrosis. Biology (Basel) 2020;9:151.ArticlePubMedPMC
  • 118. Bataller R, Schwabe RF, Choi YH, Yang L, Paik YH, Lindquist J, et al. NADPH oxidase signal transduces angiotensin II in hepatic stellate cells and is critical in hepatic fibrosis. J Clin Invest 2003;112:1383-94.ArticlePubMedPMC
  • 119. Fang PP, Pan CW, Lin W, Li J, Huang SS, Zhou GY, et al. ASK1 enhances angiotensin II-induced liver fibrosis in vitro by mediating endoplasmic reticulum stress-dependent exosomes. Mediators Inflamm 2020;2020:8183713.ArticlePubMedPMCPDF
  • 120. Wu S, Li J, Zhan Y. H3K18 lactylation accelerates liver fibrosis progression through facilitating SOX9 transcription. Exp Cell Res 2024;440:114135.ArticlePubMed
  • 121. Ogata H, Noguchi H, Ohtsubo T, Liao J, Kohara H, Yamada K, et al. Angiotensin II type I receptor blocker, losartan, inhibits fibrosis in liver by suppressing TGF-beta1 production. Integr Mol Med 2016;3:520-3.Article
  • 122. Salama ZA, Sadek A, Abdelhady AM, Darweesh SK, Morsy SA, Esmat G. Losartan may inhibit the progression of liver fibrosis in chronic HCV patients. Hepatobiliary Surg Nutr 2016;5:249-55.ArticlePubMedPMC
  • 123. Yeki M, Koda M, Matono T, Sugihara T, Maeda K, Murawaki Y. Preventative and therapeutic effects of perindopril on hepatic fibrosis induced by bile duct ligation in rats. Mol Med Rep 2009;2:857-64.PubMed

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      • PubReader PubReader
      • ePub LinkePub Link
      • Cite this Article
        Cite this Article
        export Copy Download
        Close
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        Metabolic Sparks in the Liver: Metabolic and Epigenetic Reprogramming in Hepatic Stellate Cells Activation and Its Implications for Human Metabolic Diseases
        Diabetes Metab J. 2025;49(3):368-385.   Published online May 1, 2025
        Close
      • XML DownloadXML Download
      Figure
      • 0
      • 1
      • 2
      Metabolic Sparks in the Liver: Metabolic and Epigenetic Reprogramming in Hepatic Stellate Cells Activation and Its Implications for Human Metabolic Diseases
      Image Image Image
      Fig. 1. Metabolic changes and fuel utilization in hepatic stellate cells (HSCs) activation. Quiescent HSCs play a crucial role in storing lipids and vitamins through the regulation of peroxisome proliferator-activated receptor gamma (PPARγ) and sterol regulatory element-binding protein (SREBP). During the activation into activation HSC, various signals, such as transforming growth factor-beta (TGF-β) and platelet-derived growth factor (PDGF), trigger metabolic changes to provide the energy and metabolic intermediates required for fibrogenesis. Key characteristics of this process include enhanced aerobic glycolysis, glutaminolysis, and lipolysis. Each signal is marked with a dot on the arrows where it induces metabolic changes. HGF, hepatocyte growth factor; MMP, matrix metalloproteinase; STRA6, stimulated by retinoic acid gene 6; REH, retinyl ester hydrolase; LRAT, lecithin retinol acyltransferase; SCAP, SREBP cleavage-activating protein; INSIG, insulin-induced gene; ER, endoplasmic reticulum; RE, retinyl esters; RXR, retinoid X receptor; MCT, monocarboxylate transporter; GLUT, glucose transporter; TCA, tricarboxylic acid; α-KG, α-ketoglutarate; RARβ, retinoic acid receptor β.
      Fig. 2. Metabolic reprogramming-driven epigenetic modifications in hepatic stellate cells (HSCs) activation. Intricate crosstalk between metabolic reprogramming and epigenetic modifications is essential in HSCs activation. Key metabolic alterations, including enhanced glycolysis, glutaminolysis, and loss of lipid droplets, drive histone and DNA modifications that regulate HSCs activation and fibrosis progression. Histone acetylation, lactylation, and DNA methylation are dictated by availability of metabolic intermediates such as acetyl-coenzyme A (CoA), lactate, S-adenosylmethionine (SAM), and α-ketoglutarate (α-KG). Additionally, nuclear receptor signaling modulates chromatin remodeling in response to retinoic acid and free fatty acids, thereby reinforcing transcriptional programs associated with HSCs activation. NAD+, nicotinamide adenine dinucleotide; NADH, nicotinamide adenine dinucleotide, reduced form; LDH, lactate dehydrogenase; HK2, hexokinase 2; PKM2, pyruvate M2; SIRT, sirtuin; HDAC, histone deacetylase; HAT, histone acetyltransferases; TCA, tricarboxylic acid; TET, ten-eleven translocation; CBP, CREB-binding protein; RAR, retinoic acid receptor; RXR, retinoid X receptor; NCoR, nuclear receptor corepressor; SMRT, silencer mediator of retinoid and thyroid hormone receptor; RE, retinyl esters; DNMT, DNA methyltransferase; MAT, methionine adenosyltransferase; SAH, S-adenosylhomocysteine.
      Graphical abstract
      Metabolic Sparks in the Liver: Metabolic and Epigenetic Reprogramming in Hepatic Stellate Cells Activation and Its Implications for Human Metabolic Diseases
      Causes of HSCs activation Therapeutics
      Reference
      Medications Mechanisms
      T2DM
       Hyperglycemia Metformin AMPK↑, mTOR↓, succinate-GPR91↓, electron transport chain↓ [85,86]
       Insulin resistance Thiazolidinediones (Pioglitazone) PPAR-γ↑ [87-89]
       Adipokines dysregulation SGLT2 inhibitor (Empagliflozin, Tofogliflozin) miR-34a-5p↓, hedgehog-YAP pathway↓, Hippo signaling↑ [90-92]
       AGE/RAGE signaling DPP-4 inhibitors ERK1/2, p38 and Smad2/3 phosphorylation↓, mitochondrial dysfunction↓, ROS↓ [93-95]
      GLP-1 receptor agonists RAGE/NOX2↓, ROS↓ [96,97]
      Obesity
       α-SMA/MMP9/STAT3 signaling Statin Modulating PPAR balance, Ferroptosis↑ [107,108]
       Inflamation J2H-1702 NF-κB signaling↓, Smad3/TGF-β signaling↓ [109]
       TGF-β signaling
       Lipotoxicity Adiponectin, SR1664 PPAR-γ↑ [110,111]
      Hypertension
       Oxidative stress Losartan NADPH oxidase↓, ROS↓ [115,116]
       Renin-angiotensin system Perindopril Inhibiting ACE [117]
      Table 1. Pharmacological strategies for hepatic stellate cell activation in metabolic diseases

      HSC, hepatic stellate cell; T2DM, type 2 diabetes mellitus; AGE/RAGE, advanced glycation end-products/receptor for advanced glycation endproducts; AMPK, AMP-activated protein kinase; mTOR, mechanistic target of rapamycin; GPR91, succinate-G protein-coupled receptor 91; PPAR-γ, peroxisome proliferator-activated receptor gamma; SGLT2, sodium-glucose cotransporter 2; YAP, Yes-associated protein; DPP-4, dipeptidyl peptidase-4; ERK1/2, extracellular signal-regulated kinase 1/2; ROS, reactive oxygen species; GLP-1, glucagon-like peptide-1; NOX2, NADPH oxidase 2; α-SMA, alpha-smooth muscle actin; MMP9, matrix metalloproteinase-9; STAT3, signal transducer and activator of transcription 3; TGF-β, transforming growth factor-beta; NF-κB, nuclear factor kappa B; NADPH, nicotinamide adenine dinucleotide phosphate; ACE, angiotensin-converting enzyme.

      Roh YJ, Kim H, Choi DW. Metabolic Sparks in the Liver: Metabolic and Epigenetic Reprogramming in Hepatic Stellate Cells Activation and Its Implications for Human Metabolic Diseases. Diabetes Metab J. 2025;49(3):368-385.
      Received: May 09, 2025; Accepted: Apr 17, 2025
      DOI: https://doi.org/10.4093/dmj.2025.0195.

      Diabetes Metab J : Diabetes & Metabolism Journal
      Close layer
      TOP