
Glycolysis to lactylation: Unraveling the metabolic and epigenetic landscape in tissue fibrosis (Review)
- Authors:
- Published online on: August 18, 2025 https://doi.org/10.3892/mmr.2025.13655
- Article Number: 290
-
Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Tissue fibrosis is characterized by abnormal accumulation of extracellular matrix (ECM) components, typically following persistent or repeated tissue injury (1,2). Despite its prevalence, effective therapeutic interventions to halt or reverse fibrosis remain limited, highlighting the necessity for a deeper understanding of the mechanisms driving fibrotic progression. Historically, lactate was regarded as a byproduct of glycolysis and considered metabolic waste (3,4); however, previous scientific advances have fundamentally transformed this perspective. The lactate shuttle theory has redefined lactate as a key metabolic intermediary, with its transport between cells, tissues and organs enabled through monocarboxylate transporters (MCTs) (5,6). Metabolic reprogramming, particularly enhanced glycolysis and lactate production, serves a fundamental role in fibroblast activation and ECM deposition, key characteristics of fibrotic diseases (7). Considering the diverse roles of lactate in pathological processes, particularly in tissue fibrosis, the present review provides a comprehensive examination of current knowledge regarding glycolysis, lactate metabolism and lactylation in relation to fibrosis. The present review examines how enhanced glycolysis supports the energetic and biosynthetic requirements of activated fibroblasts, how lactate functions as a signaling molecule affecting fibrogenesis and how lactylation acts as an epigenetic regulator connecting metabolism to gene expression. Through analysis of cardiac, liver, renal and pulmonary fibrosis, the present review aims to elucidate the mechanisms through which metabolic pathways contribute to fibrotic progression and discuss potential therapeutic strategies targeting these pathways.
Glycolysis and lactate metabolism
Glycolysis represents a fundamental pathway for cellular energy production (8,9). In its classic understanding, glycolysis involves glucose entering the cell via glucose transporters and undergoing conversion into two pyruvate molecules (4,10). The interconversion between lactate and pyruvate occurs through lactate dehydrogenase (LDH) via a reversible redox reaction (10). LDH exists as a tetrameric enzyme comprising two subunit types: M and H. The M subunit, encoded by the LDHA gene, predominantly exists in skeletal muscle, whereas the H subunit, encoded by the LDHB gene, primarily exists in the heart (11). LDHA primarily catalyzes pyruvate conversion to lactate, whereas LDHB promotes the reverse reaction, converting lactate to pyruvate. This structural diversity enables LDH to fulfill distinct functions across different tissues based on cellular metabolic requirements. Beyond the role of LDH, lactate exists in two enantiomeric forms in the human body: L-lactate and D-lactate (10). L-lactate constitutes the primary physiological form, predominantly produced through glycolysis, whereas D-lactate represents a minor fraction, generated through alternative metabolic pathways (12). The primary origins and pathways for L-lactate removal in the cytoplasm are illustrated in Fig. 1.
When oxygen is available, pyruvate generated in the cytoplasm can be transported into the mitochondria through the mitochondrial pyruvate carrier (MPC). Within the mitochondria, pyruvate is converted to acetyl-CoA through the enzymatic action of pyruvate dehydrogenase, which then enters the tricarboxylic acid (TCA) cycle (13,14). Considerable research has demonstrated the presence of aerobic glycolysis, wherein glucose metabolism continues to produce lactate even when oxygen levels are sufficient (15,16). While the LDH and MPC pathways are known to facilitate lactate oxidation, previous studies have highlighted the mitochondrial lactate oxidation complex, located in the inner mitochondrial membrane, as a key component in this process (17,18). In addition to its role in cellular oxidation, lactate functions as a substrate for gluconeogenesis, particularly in hepatic and skeletal muscle tissue (19). During gluconeogenesis, lactate undergoes oxidation to pyruvate before conversion to glucose, thereby maintaining glucose reserves during periods of metabolic demand (20). However, deficiencies in enzymes regulating the irreversible, rate-limiting steps of gluconeogenesis can disrupt this pathway, leading to lactate accumulation in the cytosol (Fig. 1) (21). This buildup impedes effective lactate clearance, highlighting the key role of these enzymes in maintaining metabolic balance. The lactate shuttle theory describes how lactate contributes to metabolic homeostasis through its exchange within the body via MCTs (15). MCTs, first discovered in erythrocytes, serve a key role in facilitating the transmembrane transport of pyruvate and lactate (19). These transporters are present across various cellular and organelle membranes, enabling lactate movement from regions of high to low concentration, thus supporting the lactate shuttle (22). Cells with high glycolytic activity, designated as ‘driver cells’, export lactate through MCTs, while oxidative ‘recipient cells’ import it for oxidation (6). This theory indicates that lactate functions beyond its role as a byproduct of anaerobic metabolism, serving as a notable signaling molecule within the body. This characteristic has led to the term ‘lactormone’, emphasizing the ability of lactate to regulate cellular functions through autocrine, paracrine and endocrine mechanisms (6).
Lactylation
Initially documented in 2019, lactylation represents a newly identified post-translational modification that modifies protein lysine residues using lactate molecules (23). Notably, lactylation demonstrates substantial influence on macrophage phenotypes during inflammatory responses. Research has indicated that lactylation of histone H3 at lysine 18 (H3K18la) in pro-inflammatory M1 macrophages is associated with enhanced expression of M2-like anti-inflammatory genes, indicating the essential role of lactylation in facilitating the transition from pro-inflammatory to anti-inflammatory states (23). The identification of additional proteins undergoing lactylation has increased interest into its role in tumor progression, immunity and metabolic regulation, advancing the understanding of lactate metabolism and its broader implications for cellular function (20–22).
The regulatory mechanisms of histone or non-histone protein lysine lactylation (Kla) demonstrate parallels with those of acetylation. Initially, specialized acyltransferases, designated as ‘writers’, transfer lactyl groups from L-lactyl-coenzyme A (lactyl-CoA) to lysine residues on histone or non-histone proteins, altering their structure and function. Subsequently, ‘erasers’ function as deacylases to remove these lactyl groups, terminating the Kla cycle and preventing persistent protein modifications. Finally, effector proteins, known as ‘readers’, specifically recognize the Kla modification, influencing downstream signaling pathways and initiating various biological processes (Fig. 2). Current evidence has suggested that L-lactate is converted into lactyl-CoA, facilitating the transfer of lactyl groups to lysine residues through CoA-transferases (24–26). Zhang et al (23) proposed that the enzyme p300, recognized for its acetyltransferase activity, may catalyze Kla modifications within cells. Subsequent studies have reinforced this hypothesis, demonstrating that decreased p300 expression or inhibition can lead to reduced protein lactylation (27,28). Furthermore, class I histone deacetylases 1–3 and sirtuins (SIRT) 1–3 have been identified as ‘erasers’ of histone Kla, responsible for removing lactylation markers (29,30). The glyoxalase system components also participate in lactylation, functioning as a common non-enzymatic acyl-transfer mechanism (26). During glycolysis, glyceraldehyde-3-phosphate and dihydroxyacetone phosphate are partially converted into methylglyoxal (MGO), which glyoxalase (GLO)1 subsequently transforms into lactoylglutathione (LGSH) in the presence of glutathione (31,32). GLO2 then hydrolyzes LGSH, generating D-lactate and glutathione (31). Research has indicated that MGO and its derivatives influence pathophysiological processes through D-lactylation, particularly via LGSH, which transfers lactyl groups to lysine residues, forming D-Lactoy1Lys modifications that regulate glycolysis (26). Studies have revealed that LGSH-induced D-Kla modifications are associated with enhanced inflammatory responses in macrophages (33,34). Notably, lactate-induced and LGSH-induced lactylation produce distinct outcomes. Lactate-induced lactylation promotes anti-inflammatory macrophage polarization (23), whereas LGSH-induced lactylation is associated with increased pro-inflammatory cytokine production (33). These contrasting effects suggest that the cellular role of lactylation may vary based on the underlying mechanisms. Additional research remains necessary to fully elucidate the regulatory networks governing lactate- and LGSH-mediated lactylation in cells.
Collectively, these findings indicate that lactylation functions as a bridge connecting metabolic signals to gene regulation through epigenetic modifications. Beyond protein marking, lactylation, particularly of H3K18la, reprograms transcriptional landscapes in a gene-specific and context-dependent manner. In fibrotic tissues, this regulation activates key pro-fibrotic genes including neuronal regeneration-related protein (NREP) (35) and SOX9 (36). The functional impact of lactylation varies depending on the source and type of lactate (L-lactate vs. LGSH), suggesting a sophisticated regulatory axis with both anti-and pro-inflammatory functions. Additional research is required to systematically map genome-wide lactylation patterns and identify specific reader proteins that interpret lactylation marks in fibrotic disease contexts.
Glycolysis, lactate and lactylation in tissue fibrosis
As a common end product of glycolysis, lactate exhibits multiple regulatory roles in various cell types. In this context, the distinct metabolic profiles and functions of fibroblasts, macrophages and epithelial cells during fibrosis have garnered increasing attention (37,38).
In epithelial cells, fibrotic transformation is associated with enhanced glycolysis, activation of the pentose phosphate pathway, lipid metabolic imbalance and reprogrammed amino acid metabolism (39). Mitochondrial dysfunction and accumulation of reactive oxygen species further promote epithelial-mesenchymal transition (EMT) and fibrosis (40). Additionally, epithelial-derived metabolites can activate fibroblasts (38). Fibroblasts undergo substantial metabolic reprogramming, characterized by elevated glycolysis, increased lactate production, enhanced glutamine dependence and alterations in lipid and amino acid metabolism. These metabolic shifts support fibroblast activation, proliferation and ECM deposition, while metabolite-mediated signaling amplifies fibrotic progression (41). Activated fibroblasts secrete chemokines such as chemokine (C-C motif) ligand 2 and colony stimulating factor-1, recruiting macrophages to injury sites, where they undergo polarization (42). Classically activated M1 macrophages exhibit enhanced glycolysis and pro-inflammatory responses, potentially contributing to chronic inflammation, whereas alternatively activated M2 macrophages utilize oxidative phosphorylation and fatty acid oxidation to promote tissue repair (43).
In summary, fibroblasts, macrophages and epithelial cells each demonstrate distinct metabolic changes during fibrosis, which are intricately interconnected. A comprehensive understanding of these cell-specific metabolic pathways may reveal novel therapeutic approaches for treating fibrotic diseases.
Glycolysis, lactate and lactylation in cardiac fibrosis
Cardiac fibrosis, characterized by scar formation in the heart, is a prevalent feature of various cardiac diseases (44,45). An imbalance in ECM dynamics drives fibrosis, marked by excessive accumulation of type I and III collagens (46). During fibrosis, cardiac fibroblasts (CFs) undergo activation, transforming into myofibroblasts, which increases matrix stiffness and impairs cardiac function, ultimately leading to heart failure with reduced ejection fraction (47,48). While the primary cell types within the cardiovascular system exhibit distinct metabolic characteristics in their normal state, alterations in glycolysis within fibroblasts are associated with phenotypic changes during pathological conditions (49).
Chen et al (50) used 2-deoxy-D-glucose (2-DG), a glycolysis inhibitor, to intervene in a myocardial infarction (MI)-induced fibrosis mouse model and observed a substantial reduction in collagen and α-smooth muscle actin expression levels. Additionally, clinical myocardial fibrosis samples revealed pronounced upregulation of key rate-limiting enzymes of glycolysis, including hexokinase 2 (HK2), 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) and pyruvate kinase isoform M2 (PKM2). These findings suggest that glycolysis may have a key role in post-infarction fibrosis progression. Moreover, TGF-β1-stimulated CFs, a marked elevation in extracellular acidification rate and increased lactate levels were detected (51). This indicates that CFs undergo a metabolic shift towards enhanced glycolytic activity. PFKFB3, a bifunctional isoenzyme of PFKFB with considerably increased kinase activity, serves a key role in glycolysis by synthesizing fructose-2,6-bisphosphate. A recent study demonstrated that adeno-associated virus 9-mediated gene silencing of PFKFB3 markedly suppressed fibrosis induced by MI in a mouse model (51). Another study revealed that PFKFB3 exhibits similar functions in endothelial cells by altering metabolic patterns and disrupting mitochondrial respiration, thereby promoting myocardial fibrosis (52). Based on these findings, researchers have developed a pharmacological inhibitor targeting PFKFB3, salvianolic acid C, a phenolic compound derived from Salvia miltiorrhiza. By specifically inhibiting PFKFB3-driven glycolysis, it effectively blocks endothelial-to-mesenchymal transition (EndoMT) and fibrosis progression to treat isoprenaline-induced cardiac fibrosis in a mouse model (52). Notably, previous studies have revealed that transgenic overexpression of 6-PFKFB3 in mice results in notable cardiac damage, characterized by changes in heart weight-to-body weight ratio, cardiomyocyte length and increased cardiac fibrosis (51,53). To date, several small-molecule inhibitors targeting glycolysis in myocardial fibrosis have been identified. For instance, salvianolic acid A, a phenolic compound derived from Salvia miltiorrhiza, reduces LDHA-driven glycolysis, thereby decreasing collagen-related protein (Collagen I, Collagen III) expression and ameliorating myocardial fibrosis (54). Hspa12a inhibits the activation and fibrosis of CF. It is highly expressed in resting CF and decreases after activation. Hspa12a inhibits glycolysis by stabilizing p53, blocks the transformation of CF into myofibroblasts and alleviates fibrosis after myocardial infarction. New therapeutic strategies can be developed targeting Hspa12a (55).
Beyond key glycolytic enzymes participating in myocardial fibrosis, research has demonstrated the involvement of regulators that modulate glycolysis and subsequently affect fibrosis. Chen et al (56) identified Linc00092, a long non-coding RNA involved in glycolysis, through differential screening. Further mechanistic investigations revealed that Linc00092 may regulate glycolysis-induced fibrosis in an ERK-dependent manner (56). Liu et al (57), using single-cell sequencing, identified a notable elevation in aerobic glycolysis levels in cardiac fibrosis of patients with MI. The study also revealed that the antifibrotic mechanism of IL-22 operates by regulating PKM2 via the JNK pathway (57). Another study demonstrated that the knockout of Serpina3c, a homolog of human kallistatin, exacerbates the progression of myocardial fibrosis (58). Further investigation revealed that Serpina3c interacts with Nr4a1, regulating its acetylation and promoting its degradation. This, in turn, affects enolase 1 (ENO1)-mediated glycolysis, ultimately contributing to the development of myocardial fibrosis (58).
Current research on lactate-induced lactylation in myocardial fibrosis remains limited, warranting additional investigation to increase understanding of this process. A recent study by Yao et al (59) demonstrated that elevated lactate levels can markedly accelerate cardiac fibrosis and exacerbate cardiac dysfunction by inducing endothelial-to-mesenchymal transition in the myocardium following MI.
Glycolysis, lactate and lactylation in liver fibrosis
Liver fibrosis represents a reparative response to chronic liver damage (59). As fibrosis advances, liver function deteriorates, potentially progressing to cirrhosis (60,61). At present, liver transplantation remains the sole definitive treatment for liver fibrosis (62). The fibrotic process is primarily driven by the activation of hepatic stellate cells (HSCs), which transform into myofibroblasts, generating excessive ECM components (63). Previous studies have emphasized the importance of metabolic alterations, particularly aerobic glycolysis, in facilitating HSC activation (64–66). Through glycolysis inhibition, this activation can be suppressed, as lactate serves a fundamental role in regulating gene expression levels following HSC activation. Chen et al (66) initially demonstrated that during the transformation of quiescent HSCs into myofibroblasts, glycolysis increases substantially, evidenced by enhanced expression of key glycolytic enzymes. This transformation coincides with increased intracellular lactate accumulation. The inhibition of glycolysis using 2DG or blocking lactate production with FX11 (an LDHA inhibitor) effectively regulates HSC fate, emphasizing the key role of metabolic pathways. The increased expression of these enzymes demonstrates their importance in regulating HSC activation and liver fibrosis progression (66). Subsequently, Mejias et al (64) substantiated the essential role of glycolysis in liver fibrosis progression through knockdown studies and the application of 3PO, a small molecule antagonist targeting the glycolytic rate-limiting enzyme PFKFB3. This finding received further validation in subsequent research, including studies utilizing the PKM2 antagonist shikonin and its allosteric activator TEPP-46 (67), which provided additional support for the role of glycolysis in HSC activation and liver fibrosis (68). Beyond direct involvement in HSC activation, glycolysis has been demonstrated to enhance macrophage-driven inflammatory responses, thereby intensifying fibrosis (68). These investigations provide valuable insights into the metabolic mechanisms underlying liver fibrosis and potential therapeutic targets. Given the fundamental role of glycolysis in liver fibrosis, substantial research continues to explore regulatory mechanisms and develop small molecule inhibitors targeting glycolysis.
Several key pathways and molecules have been identified: Hedgehog signaling, through transcription factor GLI family zinc finger 2, promotes glycolysis by activating hypoxia inducible factor (HIF)-1α, which controls glycolysis-related enzyme expression levels (66). Embryonic liver fordin facilitates glycolysis and ECM production in HSCs via the PI3K/Akt pathway (69). Cytoplasmic polyadenylation element binding protein 4 enhances PFKFB3 expression by binding to its untranslated mRNA region, thereby promoting glycolysis and exacerbating liver fibrosis (64). TGF-β1 triggers liver fibrosis by activating the Smad, p38 MAPK and PI3K/Akt pathways, which upregulate glucose transporter 1 (GLUT1) and increase aerobic glycolysis in HSCs (70). Wnt/β-catenin signaling activates glycolysis and HSCs by transactivating and translocating LDHA to the nucleus, where it stabilizes HIF-1α, offering a therapeutic target for fibrosis (71).
Suv39h1, a lysine methyltransferase, promotes glycolysis in HSCs by enhancing the recruitment of HIF-1α and upregulating HK2 transcription, contributing to liver fibrosis in non-alcoholic fatty liver disease (72). Focal adhesion kinase-related non-kinase inhibits aerobic glycolysis in HSCs by blocking the focal adhesion kinase/Ras/c-myc/ENO1 pathway, reducing liver fibrosis and presenting a potential therapeutic target (73). Brain and muscle arnt-like protein-1 regulates glycolysis in HSCs through the isocitrate dehydrogenase 1/α-ketoglutarate pathway, influencing liver fibrosis progression (74). Activated HSCs have been demonstrated to transfer GLUT1 and PKM2 to quiescent HSCs, Kupffer cells and liver sinusoidal endothelial cells via exosomes, further exacerbating liver fibrosis (75). Glycolysis amplifies liver fibrosis by promoting the release of fibrogenic extracellular vesicles (EVs) from HSCs, particularly in the hepatic pericentral zone, highlighting glycolysis as a therapeutic target (76). Additionally, several natural compounds have demonstrated potential in targeting glycolysis and mitigating fibrosis: Curcumin has been shown to alleviate liver fibrosis by modulating Hedgehog signaling activation and adenosine monophosphate-activated protein kinase (AMPK), thereby influencing glycolysis (77,78). The natural compound oroxylin A has been demonstrated to inhibit both the expression and activity of LDHA in HSCs, suggesting its potential as a therapeutic agent in reducing glycolysis and mitigating liver fibrosis (79). Costunolide has been reported to target HK2, thereby disrupting the glycolytic process, inhibiting the activation of HSCs and slowing the progression of liver fibrosis (80). Furthermore, various other small molecules that regulate glycolysis have been discovered, highlighting glycolysis as a potential therapeutic target for liver fibrosis (81–86).
Recent research has revealed that lactylation, driven by glycolysis-produced lactate, may be a key factor in HSC activation (87). Studies have demonstrated that lactate generated by HK2 in HSCs promotes histone lactylation, including modifications such as H3K18la, H3K9la, H3K14la, H4K8la and H4K12la (87,88). These lactylation modifications influence gene expression, thereby contributing to HSC activation and liver fibrosis progression (87). Furthermore, insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2), an m6A-binding protein, has been identified as promoting liver fibrosis through regulation of glycolysis and aldolase A (ALDOA) expression levels, resulting in histone lactylation and HSC activation, suggesting its potential as a therapeutic target (89). Notably, H3K18la enhances SOX9 transcription, a key fibrosis factor, indicating that this pathway may present novel therapeutic opportunities for hepatic fibrosis treatment (90). Rho et al (87) established that HK2-driven lactate production in HSCs modulates gene expression levels through histone lactylation. Their research revealed that HSC activation-related gene promoters demonstrate H3K18la enrichment and increased glycolysis results in widespread lactylation of lysine residues on both histone H3 and H4 and non-histone proteins. Additionally, IGF2BP2, an m6A reader protein that enhances mRNA stability, has been revealed to be notably upregulated in liver fibrosis, whereas IGF2BP2 suppression can attenuate fibrotic progression. The mechanism involves IGF2BP2 enhancing the m6A modification and stability of ALDOA mRNA, leading to increased ALDOA expression levels and lactate production, which subsequently promotes H3K18la and accelerates liver fibrosis (91). Wu et al (90) revealed that during liver fibrosis, the lactation level of H3K18 increases, and when LDHA is reduced, H3K18 lactation decreases, inhibiting the activation and fibrotic process of HSC. Further experiments demonstrated that H3K18 lactation promotes liver fibrosis by enhancing SOX9 gene expression, and overexpression of SOX9 can reverse the anti-fibrotic effect of reduced LDHA, indicating that H3K18 lactation promotes liver fibrosis development by regulating SOX9 (90).
Chen et al (92) revealed that lactate-driven H3K18la upregulates interferon regulatory factor 4 transcription, promotes the differentiation of CD4 T cells into T helper 17 cells and leads to an increase in IL-17A secretion, thereby accelerating the activation of HSCs and exacerbating arsenite-induced liver fibrosis. Zhou et al (89) revealed that in a mouse model of carbon tetrachloride-induced liver fibrosis, the overall lactation level increased, and in TGF-β1-treated LX-2 cells with liver fibrosis, both the overall lactation and H3K18la levels increased. By contrast, correcting abnormal lactation or supplementing lactic acid may alter the activated phenotype of LX-2 cells, while interfering with lactic acid production reduces H3K18la levels and inactivates HSCs (89). Moreover, as H3K18la is enriched in activated HSCs, these findings suggest that H3K18la is associated with liver fibrosis (89). However, the current understanding of lactylation modifications in liver fibrosis remains incomplete, necessitating further research to fully understand their roles in fibrogenesis.
Glycolysis, lactate and lactylation in renal fibrosis
Chronic kidney disease (CKD) represents a notable public health challenge (93). Renal fibrosis, characterized by excessive ECM accumulation in the interstitial space of the kidneys, is a defining feature of CKD (94,95). Emerging evidence has increasingly demonstrated the relationship between glycolysis and renal fibrosis. The importance of glycolysis in renal fibrosis was initially demonstrated by Ding et al (96); this previous study revealed that aerobic glycolysis inhibitors, such as 2-DG, or PKM gene silencing with its inhibitor shikonin, effectively reduced fibrosis in a mouse model of unilateral ureteral obstruction and TGF-β-induced fibroblast activation. These observations have been supported by studies demonstrating that PKM2 overexpression in rat renal fibroblasts can directly trigger fibroblast activation and enhance glycolytic activity (97,98). PKM2, a key glycolytic enzyme, exists in multiple forms, monomers, dimers and tetramers, each serving distinct pathophysiological functions (99). Liu et al (100) demonstrated that TEPP-46, a small-molecule activator that stabilizes PKM2 tetramer formation and enhances PK activity, inhibited both renal fibrosis and glycolysis in kidney cells (101,102). Further research by Wei et al (103) and Yu et al (104) revealed that glycolysis inhibition using dichloroacetate (DCA) (103) or 3-bromopyruvate (104) could reduce renal fibrosis. Notably, the study by Wei et al (103) indicated that glycolysis may have varying effects in different cell types, while glycolysis inhibition can reduce fibrosis in fibroblasts, it may promote fibrosis in renal tubular epithelial cells.
Transgenic models have further validated the essential role of glycolysis in renal fibrosis. Lee et al (105) demonstrated that PFKFB2 isoform phosphorylation in the kidney markedly influences fibrosis progression. Mutations inactivating Ser468 and Ser485 of PFKFB2 (PFKFB2 knock-in mice) have been shown to intensify renal fibrosis (105), whereas specific PFKFB2 knockout in renal fibroblasts (106), myeloid cells (107) and kidney proximal tubular cells (108) can reduce fibrosis progression.
The regulatory mechanisms underlying glycolysis in renal fibrosis have emerged as a central focus of research, particularly in developing small-molecule inhibitors targeting this metabolic process. Srivastava et al (109) demonstrated that SIRT3 deficiency contributes to diabetes-associated kidney fibrosis through mechanisms involving HIF-1α accumulation and PKM2 dimer formation. Research has also indicated that Honokiol-induced activation of SIRT3 mitigates fibrosis, potentially through deacetylation of pyruvate dehydrogenase E1α, an enzyme connecting glycolysis to the TCA cycle (110). Furthermore, aldehyde dehydrogenase 2 has been identified as a notable factor in glycolysis-induced renal fibrosis (111,112). Tuberous sclerosis complex 1 (Tsc1)-mediated target of rapamycin complex 1 activation contributes to the glycolytic process in renal tubular cells, thereby intensifying fibrosis (113). Various other molecules have been demonstrated to be involved in glycolysis-mediated fibrosis, including Tsc1 (113), COUP-TFII (NR2F2) (114), GPR87 (115), YY1 (116), platelet isoform of PFK (117) and forkhead box protein K1 (118). Additionally, signaling pathways such as the cGAS-STING pathway increase PFKFB3 expression levels, contributing to hypoxia-induced renal fibrosis (119). HIF-1α has a key role in glycolysis regulation. Both Sanqi oral solution (120) and sodium-glucose cotransporter 2 (SGLT2) inhibitors (121) demonstrate effectiveness in modulating HIF-1α expression levels, thereby reducing glycolysis and its fibrogenic effects. Traditional Chinese medicine formulations, including Huangqi-Danshen decoction (122) and Shen Shuai II Recipe (123), have also been demonstrated to exhibit efficacy in regulating glycolysis and diminishing fibrosis progression.
Recent studies have revealed that lactylation, driven by lactate produced through glycolysis, may constitute a key factor in the progression of renal fibrosis (108,115). Wang et al (108) demonstrated that persistent glycolysis in kidney tubular epithelial cells promotes histone H4 lysine 12 lactylation, activating NF-κB-related gene transcription, thus establishing a connection between metabolism and inflammation-mediated fibrosis in renal fibrosis. Despite these advances, the study of lactylation modifications in renal fibrosis requires additional research to elucidate their specific contributions to fibrogenesis.
Glycolysis, lactate and lactylation in idiopathic pulmonary fibrosis (IPF)
IPF is characterized by excessive accumulation of ECM proteins, resulting in progressive lung tissue scarring and compromised respiratory function. Research has emphasized the fundamental role of metabolic reprogramming, particularly enhanced glycolysis, in IPF pathogenesis. Xie et al (124) identified glycolytic reprogramming as an essential early event in myofibroblast differentiation. Their research demonstrated elevated levels of enzymes including PFK1, HK2 and PFKFB3 in fibrotic lungs, leading to increased glycolysis. Supporting these findings, Kang et al (125) utilized metabolic profiling to reveal elevated levels of glycolysis-related metabolites in IPF lung tissues, indicating a shift toward aerobic glycolysis. Zhao et al (126) revealed altered glycolytic intermediates in IPF lungs, characterized by reduced levels of key glycolytic enzymes and increased lactic acid, suggesting disrupted glycolysis and a shift toward anaerobic metabolism. Notably, GLUT1 demonstrates considerable importance in this metabolic shift. Cho et al (127) revealed elevated GLUT1-dependent glycolysis in age-associated lung fibrosis. By contrast, GLUT1 inhibition using phloretin reduced fibrotic markers and improved lung function in mice, highlighting GLUT1 as a potential therapeutic target (127).
Key glycolytic enzymes serve a notable role in fibroblast activation in IPF. Kim et al (128) demonstrated that HK2 connects glycolysis to the profibrotic effects of TGF-β (129), and HK2 inhibition can decrease collagen production and myofibroblast differentiation (129). Hu et al (130) established that the PI3K-Akt-mTOR/PFKFB3 pathway controls glycolysis and collagen production in lung fibroblasts during lipopolysaccharide-induced pulmonary fibrosis. Notably, PFKFB3 inhibition can reduce glycolytic flux and collagen deposition (130). Research has also indicated that metformin, a common antidiabetic drug, inhibits PFKFB3-mediated glycolysis and decreases collagen production in lung fibroblasts through modulation of the AMPK/mTOR pathway (131). Similarly, Chen et al (132) determined that anlotinib suppresses PFKFB3-driven glycolysis in myofibroblasts, reversing pulmonary fibrosis in bleomycin-induced mouse models. The mTORC1 pathway also influences metabolic reprogramming in IPF. O'Leary et al (133) revealed that TGF-β drives metabolic reprogramming in lung fibroblasts by triggering activating transcription factor 4 through an mTORC1-dependent mechanism, enhancing serine and glycine synthesis essential for collagen production. HIF-1α serves as another key regulator of glycolysis in IPF. Xu et al (134) established that high mobility group box protein 1 promotes fibroblast proliferation and ECM production through activating HIF1α-regulated aerobic glycolysis. Goodwin et al (135) revealed that targeting the HIF1α/3-phosphoinositide-dependent protein kinase 1 (PDK1) axis with DCA inhibits bleomycin-induced pulmonary fibrosis by suppressing glycolysis. Additionally, the Akt2-PDK1 signaling pathway contributes to lactate accumulation and fibrosis. Sun et al (136) demonstrated that lactate buildup, driven by Akt2-PDK1 signaling, promotes pulmonary fibrosis through increased glycolysis. By contrast, Schruf et al (137) theorized that the fibroblast-to-myofibroblast transformation does not result from an LDH5-dependent metabolic shift towards aerobic glycolysis, indicating that glycolysis might not be the sole driver of fibrosis. In mechanical ventilation-induced pulmonary fibrosis, Mei et al (138) revealed that the integrin β3-PKM2 pathway mediates aerobic glycolysis, contributing to fibrosis. In addition, it has been demonstrated that fenbendazole attenuates fibrosis by inhibiting glycolysis and fibroblast activation (139). Furthermore, the alamandine/MrgD axis prevents TGF-β1-mediated fibroblast activation by regulating aerobic glycolysis and promoting mitophagy (140). Dishevelled-binding antagonist of β-catenin 2 protects against pulmonary fibrosis by suppressing glycolysis in lung myofibroblasts (141).
Lactate has emerged as a key signaling molecule in IPF. Cui et al (28) revealed that lung myofibroblasts enhance macrophage profibrotic activity through lactate-induced histone lactylation. Histone lactylation represents a novel epigenetic modification connecting cellular metabolism to gene expression. Wang et al (35) demonstrated that H3K18la promotes the progression of arsenite-related IPF via the YTH domain-containing family protein 1/m6A/NREP pathway. Arsenite exposure increases lactate production, resulting in enhanced histone lactylation and activation of profibrotic signaling pathways (35). Li et al (142) revealed that airborne PM2.5 induces pulmonary fibrosis by activating glycolysis and triggering histone lactylation in macrophages. The produced lactate enhances EMT in epithelial cells, promoting fibrosis (142). Feng et al (143) revealed that lipopolysaccharide-induced inhibition of monocarboxylate transporter 1 (MCT1) leads to lactate accumulation, initiating EMT and pulmonary fibrosis. Enhancement of lactate transport through MCT1 reduces lactate levels and mitigates fibrosis (143). Additionally, lactate serves a role in intercellular communication in the fibrotic microenvironment. Wang et al (135) revealed that microRNA-21 in EVs from pulmonary epithelial cells promotes myofibroblast differentiation via glycolysis in arsenic-induced pulmonary fibrosis. Lactate accumulation increases glycolytic activity in fibroblasts, driving their activation, thereby creating a feed-forward loop that exacerbates fibrosis.
In summary, enhanced glycolysis in lung fibroblasts serves a key role in the pathogenesis of IPF by providing energy and biosynthetic precursors essential for fibroblast activation and ECM production. Key glycolytic enzymes and pathways, including GLUT1, HK2, PFKFB3, and the mTORC1 and HIF-1α pathways, demonstrate upregulation in IPF and present potential therapeutic targets (114,117–119,122). Lactate, the end product of glycolysis, functions not merely as a metabolic byproduct but as a signaling molecule that promotes fibrosis through histone lactylation and modulation of gene expression (28,35). Lactate-induced histone lactylation in macrophages and epithelial cells augments the expression of profibrotic genes and facilitates processes such as EMT, contributing to fibrosis progression. Targeting metabolic pathways involved in glycolysis and lactate metabolism presents promising therapeutic strategies for IPF. Interventions including glycolysis inhibitors, lactate transport modulators and agents that disrupt lactate signaling demonstrate efficacy in preclinical models (131,132,134,135,141,143). Understanding the complex relationship between cellular metabolism and fibrotic signaling pathways remains essential for developing novel treatments for IPF. Future research should emphasize on translating these findings into clinical therapies that can halt or reverse fibrosis by targeting metabolic dysregulation.
Future perspective
The emerging understanding of glycolysis, lactate metabolism and lactylation in tissue fibrosis reveals new avenues for research and therapeutic intervention (Fig. 3). Despite considerable progress, several key questions remain unanswered. Firstly, while the role of enhanced glycolysis in fibroblast activation is well-documented, the precise regulatory mechanisms governing this metabolic shift require further investigation. Examining the upstream signals that initiate metabolic reprogramming in fibroblasts and other cell types within the fibrotic microenvironment could identify novel targets for intervention. Additionally, understanding how different cell types interact metabolically during fibrosis might reveal important intercellular communication pathways mediated by metabolites such as lactate. Secondly, the importance of lactate as a signaling molecule and its role in histone lactylation warrant deeper exploration. Future studies should aim to map the complete spectrum of proteins subject to lactylation in fibrotic tissues, determine the functional consequences of these modifications, and identify the enzymes involved in writing, reading and erasing lactylation markers. Glycolysis inhibitors, lactate transport modulators and agents affecting lactylation enzymes present potential strategies. HIF-1α, a glycolytic master transcriptional regulator that transactivates several glycolytic genes, is upregulated in IPF and represents a potential target for fibrosis treatment (144,145). Hu et al (120) demonstrated that Sanqi oral solution can inhibit the HIF-1α/PKM2/glycolytic-fibroblast activation axis and delay renal fibrosis progression. LDHA primarily functions to convert pyruvate into lactic acid. Salvianolic acid A has demonstrated inhibition of LDHA-driven glycolysis, thereby reducing fibrosis severity (54). HSC activation represents a target for hepatic fibrosis, with aerobic glycolysis as one of its metabolic characteristics; therefore, blocking glycolysis presents a new therapeutic direction for hepatic fibrosis (146). Ban et al (80) revealed that costunolide can inhibit HK2, reduce HSC glycolysis and thus inhibit the activation of HSCs. However, given the essential roles of these metabolic processes in normal physiology, specificity and minimizing off-target effects remain key. The development of targeted delivery systems or prodrugs that preferentially accumulate in fibrotic tissues may enhance therapeutic efficacy while reducing systemic toxicity.
Emerging technologies such as single-cell RNA sequencing, metabolomics and advanced imaging techniques could prove instrumental in dissecting the complex metabolic networks in fibrosis. Integration of these data could facilitate the identification of metabolic signatures predictive of disease progression or therapeutic response, establishing foundations for personalized medicine approaches in fibrosis management.
Conclusion
In conclusion, the complex interplay between glycolysis, lactate metabolism and lactylation substantially contributes to the pathogenesis of tissue fibrosis. Enhanced glycolysis provides the essential energy and biosynthetic precursors for activated fibroblasts, while lactate functions as a signaling molecule influencing fibroblast activation, macrophage polarization and EMT. The discovery of lactylation as a novel epigenetic modification emphasizes the profound impact of metabolic changes on gene expression regulation during fibrosis. Understanding these metabolic and epigenetic mechanisms provides new insights into fibrotic disease progression and highlights potential therapeutic targets. Interventions aimed at modulating glycolysis, lactate production and lactylation demonstrate promise in preclinical models, suggesting metabolic reprogramming as a viable strategy for treating fibrosis.
Future research should focus on elucidating the detailed molecular mechanisms, exploring the specificity of potential therapeutic agents and translating these findings into clinical applications. Through continued investigation of the metabolic and epigenetic landscape of fibrosis, progress continues toward developing effective therapies that can improve outcomes for patients affected by these conditions.
Acknowledgements
Not applicable.
Funding
The present study was supported by the National Natural Science Foundation of China (grant no. 82104665); The Science and Technology Department of Sichuan Province (grant nos. 2023NSFSC1763 and 2022YFS0621); The Innovation Team Project of Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University (grant no. 2022-CXTD-03); The College Students Innovation and Entrepreneurship Training Program (grant no. 202410632017); and The Science and Technology Strategic Cooperation Programs of Luzhou Municipal People's Government and Southwest Medical University (grant no. 2024LZXNYKDJ020).
Availability of data and materials
Not applicable.
Authors' contributions
LZ collected the literature and wrote the manuscript. QL and YD contributed to literature collection, and analysis and interpretation of the results. LZ, YZ, LW and JL revised the manuscript. All authors reviewed the final draft of the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Yu XY, Sun Q, Zhang YM, Zou L and Zhao YY: TGF-β/Smad signaling pathway in tubulointerstitial fibrosis. Front Pharmacol. 13:8605882022. View Article : Google Scholar : PubMed/NCBI | |
Zhao X, Kwan JYY, Yip K, Liu PP and Liu FF: Targeting metabolic dysregulation for fibrosis therapy. Nat Rev Drug Discovery. 19:57–75. 2020. View Article : Google Scholar : PubMed/NCBI | |
Vavřička J, Brož P, Follprecht D, Novák J and Kroužecký A: Modern perspective of lactate metabolism. Physiol Res. 73:499–514. 2024. View Article : Google Scholar : PubMed/NCBI | |
Rabinowitz JD and Enerbäck S: Lactate: The ugly duckling of energy metabolism. Nat Metab. 2:566–571. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ren LL, Miao H, Wang YN, Liu F, Li P and Zhao YY: TGF-β as A master regulator of Aging-associated tissue fibrosis. Aging Dis. 14:1633–1650. 2023. View Article : Google Scholar : PubMed/NCBI | |
Brooks GA: The science and translation of lactate shuttle theory. Cell Metab. 27:757–785. 2018. View Article : Google Scholar : PubMed/NCBI | |
Miguel V, Alcalde-Estévez E, Sirera B, Rodríguez-Pascual F and Lamas S: Metabolism and bioenergetics in the pathophysiology of organ fibrosis. Free Radic Biol Med. 222:85–105. 2024. View Article : Google Scholar : PubMed/NCBI | |
Pucino V, Bombardieri M, Pitzalis C and Mauro C: Lactate at the crossroads of metabolism, inflammation, and autoimmunity. Eur J Immunol. 47:14–21. 2017. View Article : Google Scholar : PubMed/NCBI | |
Maciolek JA, Alex Pasternak J and Wilson HL: Metabolism of activated T lymphocytes. Curr Opin Immunol. 27:60–74. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li X, Yang Y, Zhang B, Lin X, Fu X, An Y, Zou Y, Wang JX, Wang Z and Yu T: Lactate metabolism in human health and disease. Signal Transduct Target Ther. 7:3052022. View Article : Google Scholar : PubMed/NCBI | |
Urbańska K and Orzechowski A: Unappreciated role of LDHA and LDHB to control apoptosis and autophagy in tumor cells. Int J Mol Sci. 20:20852019. View Article : Google Scholar : PubMed/NCBI | |
Dawson DM, Goodfriend TL and Kaplan NO and Kaplan NO: Lactic Dehydrogenases: Functions of the two types: Rates of synthesis of the two major forms can be correlated with metabolic differentiation. Science. 143:929–933. 1964. View Article : Google Scholar : PubMed/NCBI | |
Martínez-Reyes I and Chandel NS: Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 11:1022020. View Article : Google Scholar : PubMed/NCBI | |
Gray LR, Tompkins SC and Taylor EB: Regulation of pyruvate metabolism and human disease. Cell Mol Life Sci. 71:2577–2604. 2014. View Article : Google Scholar : PubMed/NCBI | |
Brooks GA, Arevalo JA, Osmond AD, Leija RG, Curl CC and Tovar AP: Lactate in contemporary biology: A phoenix Risen. J Physiol. 600:1229–1251. 2022. View Article : Google Scholar : PubMed/NCBI | |
Brooks GA: Lactate as a fulcrum of metabolism. Redox Biol. 35:1014542020. View Article : Google Scholar : PubMed/NCBI | |
Brooks GA, Curl CC, Leija RG, Osmond AD, Duong JJ and Arevalo JA: Tracing the lactate shuttle to the mitochondrial reticulum. Exp Mol Med. 54:1332–1347. 2022. View Article : Google Scholar : PubMed/NCBI | |
Leija RG, Arevalo JA, Xing D, Vázquez-Medina JP and Brooks GA: The mitochondrial lactate oxidation complex: Endpoint for carbohydrate carbon disposal. Am J Physiol Endocrinol Metab. 328:E126–E136. 2025. View Article : Google Scholar : PubMed/NCBI | |
Zhu W, Guo S, Sun J, Zhao Y and Liu C: Lactate and lactylation in cardiovascular diseases: Current progress and future perspectives. Metabolis. 158:1559572024. View Article : Google Scholar : PubMed/NCBI | |
Stumvoll M, Meyer C, Mitrakou A, Nadkarni V and Gerich JE: Renal glucose production and utilization: New aspects in humans. Diabetologia. 40:749–757. 1997. View Article : Google Scholar : PubMed/NCBI | |
Li H, Ren Q, Shi M, Ma L and Fu P: Lactate metabolism and acute kidney injury. Chin Med J (Engl). 138:916–924. 2025. View Article : Google Scholar : PubMed/NCBI | |
Jones R and Morris M: Monocarboxylate transporters: Therapeutic targets and prognostic factors in disease. Clin Pharmacol Ther. 100:454–463. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, et al: Metabolic regulation of gene expression by histone lactylation. Nature. 574:575–580. 2019. View Article : Google Scholar : PubMed/NCBI | |
Xiong J, He J, Zhu J, Pan J, Liao W, Ye H, Wang H, Song Y, Du Y, Cui B, et al: Lactylation-driven METTL3-mediated RNA m6A modification promotes immunosuppression of Tumor-infiltrating myeloid cells. Mol Cell. 82:1660–1677.e10. 2022. View Article : Google Scholar : PubMed/NCBI | |
Yang K, Fan M, Wang X, Xu J, Wang Y, Tu F, Gill PS, Ha T, Liu L, Williams DL and Li C: Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Cell Death Differ. 29:133–146. 2022. View Article : Google Scholar : PubMed/NCBI | |
Gaffney DO, Jennings EQ, Anderson CC, Marentette JO, Shi T, Schou Oxvig AM, Streeter MD, Johannsen M, Spiegel DA, Chapman E, et al: Non-enzymatic lysine lactoylation of glycolytic enzymes. Cell Chem Biol. 27:206–213.e6. 2020. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Zhang Y, Li W and Zhou X: Lactylation, an emerging hallmark of metabolic reprogramming: Current progress and open challenges. Front Cell Dev Biol. 10:9720202022. View Article : Google Scholar : PubMed/NCBI | |
Cui H, Xie N, Banerjee S, Ge J, Jiang D, Dey T, Matthews QL, Liu RM and Liu G: Lung myofibroblasts promote macrophage profibrotic activity through Lactate-induced histone lactylation. Am J Respir Cell Mol Biol. 64:115–125. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wei S, Gao Y, Dai X, Fu W, Cai S, Fang H, Zeng Z and Chen Z: SIRT1-mediated HMGB1 deacetylation suppresses sepsis-associated acute kidney injury. Am J Physiol Renal Physiol. 316:F20–F31. 2019. View Article : Google Scholar : PubMed/NCBI | |
Varner EL, Trefely S, Bartee D, von Krusenstiern E, Izzo L, Bekeova C, O'Connor RS, Seifert EL, Wellen KE, Meier JL and Snyder NW: Quantification of lactoyl-CoA (lactyl-CoA) by liquid chromatography mass spectrometry in mammalian cells and tissues. Open Biol. 10:2001872020. View Article : Google Scholar : PubMed/NCBI | |
Rabbani N, Xue M and Thornalley PJ: Activity, regulation, copy number and function in the glyoxalase system. Biochem Soc Trans. 42:419–424. 2014. View Article : Google Scholar : PubMed/NCBI | |
Allaman I, Bélanger M and Magistretti PJ: Methylglyoxal, the dark side of glycolysis. Front Neurosci. 9:232015. View Article : Google Scholar : PubMed/NCBI | |
Trujillo MN, Jennings EQ, Hoffman EA, Zhang H, Phoebe AM, Mastin GE, Kitamura N, Reisz JA, Megill E, Kantner D, et al: Lactoylglutathione promotes inflammatory signaling in macrophages through histone lactoylation. Mol Metab. 81:1018882024. View Article : Google Scholar : PubMed/NCBI | |
Dichtl S, Lindenthal L, Zeitler L, Behnke K, Schlösser D, Strobl B, Scheller J, El Kasmi KC and Murray PJ: Lactate and IL6 define separable paths of inflammatory metabolic adaptation. Sci Adv. 7:eabg35052021. View Article : Google Scholar : PubMed/NCBI | |
Wang P, Xie D, Xiao T, Cheng C, Wang D, Sun J, Wu M, Yang Y, Zhang A and Liu Q: H3K18 lactylation promotes the progression of arsenite-related idiopathic pulmonary fibrosis via YTHDF1/m6A/NREP. J Hazard Mater. 461:1325822024. View Article : Google Scholar : PubMed/NCBI | |
Aggarwal S, Wang Z, Rincon Fernandez Pacheco D, Rinaldi A, Rajewski A, Callemeyn J, Van Loon E, Lamarthée B, Covarrubias AE, Hou J, et al: SOX9 switch links regeneration to fibrosis at the single-cell level in mammalian kidneys. Science. 383:eadd63712024. View Article : Google Scholar : PubMed/NCBI | |
Wynn TA and Vannella KM: Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 44:450–462. 2016. View Article : Google Scholar : PubMed/NCBI | |
Marconi GD, Fonticoli L, Rajan TS, Pierdomenico SD, Trubiani O, Pizzicannella J and Diomede F: Epithelial-mesenchymal transition (EMT): The Type-2 EMT in wound healing, tissue regeneration and organ fibrosis. Cells. 10:15872021. View Article : Google Scholar : PubMed/NCBI | |
Tang W and Wei Q: The metabolic pathway regulation in kidney injury and repair. Front Physiol. 14:13442712024. View Article : Google Scholar : PubMed/NCBI | |
Richter K and Kietzmann T: Reactive oxygen species and fibrosis: Further evidence of a significant liaison. Cell Tissue Res. 365:591–605. 2016. View Article : Google Scholar : PubMed/NCBI | |
Feng L, Chen X, Huang Y, Zhang X, Zheng S and Xie N: Immunometabolism changes in fibrosis: From mechanisms to therapeutic strategies. Front Pharmacol. 14:12436752023. View Article : Google Scholar : PubMed/NCBI | |
Liu SS, Liu C, Lv XX, Cui B, Yan J, Li YX, Li K, Hua F, Zhang XW, Yu JJ, et al: The chemokine CCL1 triggers an AMFR-SPRY1 pathway that promotes differentiation of lung fibroblasts into myofibroblasts and drives pulmonary fibrosis. Immunity. 54:2042–2056.e8. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen S, Wang K, Fan Z, Zhou T, Li R, Zhang B, Chen J, Chi J, Wei K, Liu J, et al: Modulation of anti-cardiac fibrosis immune responses by changing M2 macrophages into M1 macrophages. Mol Med. 30:882024. View Article : Google Scholar : PubMed/NCBI | |
Weber KT, Sun Y, Bhattacharya SK, Ahokas RA and Gerling IC: Myofibroblast-mediated mechanisms of pathological remodelling of the heart. Nat Rev Cardiol. 10:15–26. 2013. View Article : Google Scholar : PubMed/NCBI | |
Segura AM, Frazier OH and Buja LM: Fibrosis and heart failure. Heart Fail Rev. 19:173–185. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li L, Zhao Q and Kong W: Extracellular matrix remodeling and cardiac fibrosis. Matrix Biol. 68-69:490–506. 2018. View Article : Google Scholar : PubMed/NCBI | |
Disertori M, Masè M and Ravelli F: Myocardial fibrosis predicts ventricular tachyarrhythmias. Trends Cardiovasc Med. 27:363–372. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu M, López De Juan Abad B and Cheng K: Cardiac fibrosis: Myofibroblast-mediated pathological regulation and drug delivery strategies. Adv Drug Delivery Rev. 173:504–519. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gibb AA, Lazaropoulos MP and Elrod JW: Myofibroblasts and fibrosis: Mitochondrial and metabolic control of cellular differentiation. Circ Res. 127:427–447. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chen ZT, Gao QY, Wu MX, Wang M, Sun RL, Jiang Y, Guo Q, Guo DC, Liu CY, Chen SX, et al: Glycolysis inhibition alleviates cardiac fibrosis after myocardial infarction by suppressing cardiac fibroblast activation. Front Cardiovasc Med. 8:7017452021. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Yin X, Fan YM, Zhang X, Ma C, Jia K, Zhou W, Tang Z, Qi LW and Li J: Upregulation of glycolytic enzyme PFKFB3 by deubiquitinase OTUD4 promotes cardiac fibrosis post myocardial infarction. J Mol Med. 101:743–756. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zeng H, Pan T, Zhan M, Hailiwu R, Liu B, Yang H and Li P: Suppression of PFKFB3-driven glycolysis restrains endothelial-to-mesenchymal transition and fibrotic response. Signal Transduct Target Ther. 7:3032022. View Article : Google Scholar : PubMed/NCBI | |
Wang Q, Donthi RV, Wang J, Lange AJ, Watson LJ, Jones SP and Epstein PN: Cardiac phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase increases glycolysis, hypertrophy, and myocyte resistance to hypoxia. Am J Physiol Heart Circ Physiol. 294:H2889–H2897. 2008. View Article : Google Scholar : PubMed/NCBI | |
Hailiwu R, Zeng H, Zhan M, Pan T, Yang H and Li P: Salvianolic acid A diminishes LDHA-driven aerobic glycolysis to restrain myofibroblasts activation and cardiac fibrosis via blocking Akt/GSK-3β/HIF-1α axis. Phytother Res. 37:4540–4556. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mao Q, Zhang X, Yang J, Kong Q, Cheng H, Yu W, Cao X, Li Y, Li C, Liu L and Ding Z: HSPA12A acts as a scaffolding protein to inhibit cardiac fibroblast activation and cardiac fibrosis. J Adv Res. 67:217–229. 2025. View Article : Google Scholar : PubMed/NCBI | |
Chen ZT, Zhang HF, Wang M, Wang SH, Wen ZZ, Gao QY, Wu MX, Liu WH, Xie Y, Mai JT, et al: Long non-coding RNA Linc00092 inhibits cardiac fibroblast activation by altering glycolysis in an ERK-dependent manner. Cell Signal. 74:1097082020. View Article : Google Scholar : PubMed/NCBI | |
Liu F, Chen Y, Qin D and Qian C: Interleukin-22 inhibits cardiac fibrosis by regulating fibroblast metabolic reprogramming in myocardial infarction. Pathol Res Pract. 256:1552562024. View Article : Google Scholar : PubMed/NCBI | |
Ji J, Qian L, Zhu Y, Jiang Y, Guo JQ, Wu Y, Yang ZW, Yao YY and Ma GS: Kallistatin/Serpina3c inhibits cardiac fibrosis after myocardial infarction by regulating glycolysis via Nr4a1 activation. Biochim Biophys Acta Mol Basis Dis. 1868:1664412022. View Article : Google Scholar : PubMed/NCBI | |
Yao S, Yang X, An J, Jin H, Wen G, Wang H and Tuo B: Role of the S100 protein family in liver disease (Review). Int J Mol Med. 48:1662021. View Article : Google Scholar : PubMed/NCBI | |
Roehlen N, Crouchet E and Baumert TF: Liver fibrosis: Mechanistic concepts and therapeutic perspectives. Cells. 9:8752020. View Article : Google Scholar : PubMed/NCBI | |
Dulai PS, Singh S, Patel J, Soni M, Prokop LJ, Younossi Z, Sebastiani G, Ekstedt M, Hagstrom H, Nasr P, et al: Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology. 65:1557–1565. 2017. View Article : Google Scholar : PubMed/NCBI | |
Tan Z, Sun H, Xue T, Gan C, Liu H, Xie Y, Yao Y and Ye T: Liver fibrosis: Therapeutic Targets and advances in drug therapy. Front Cell Dev Biol. 9:7301762021. View Article : Google Scholar : PubMed/NCBI | |
Yao S, Chai H, Tao T, Zhang L, Yang X, Li X, Yi Z, Wang Y, An J, Wen G, et al: Role of lactate and lactate metabolism in liver diseases (Review). Int J Mol Med. 54:592024. View Article : Google Scholar : PubMed/NCBI | |
Mejias M, Gallego J, Naranjo-Suarez S, Ramirez M, Pell N, Manzano A, Suñer C, Bartrons R, Mendez R and Fernandez M: CPEB4 increases expression of PFKFB3 to induce glycolysis and activate mouse and human hepatic stellate cells, promoting liver fibrosis. Gastroenterology. 159:273–288. 2020. View Article : Google Scholar : PubMed/NCBI | |
Trivedi P, Wang S and Friedman SL: The power of Plasticity-metabolic regulation of hepatic stellate cells. Cell Metab. 33:242–257. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Choi SS, Michelotti GA, Chan IS, Swiderska-Syn M, Karaca GF, Xie G, Moylan CA, Garibaldi F, Premont R, et al: Hedgehog controls hepatic stellate cell fate by regulating metabolism. Gastroenterology. 143:1319–1329.e11. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zheng D, Jiang Y, Qu C, Yuan H, Hu K, He L, Chen P, Li J, Tu M, Lin L, et al: Pyruvate kinase M2 tetramerization protects against hepatic stellate cell activation and liver fibrosis. Am J Pathol. 190:2267–2281. 2020. View Article : Google Scholar : PubMed/NCBI | |
Rao J, Wang H, Ni M, Wang Z, Wang Z, Wei S, Liu M, Wang P, Qiu J, Zhang L, et al: FSTL1 promotes liver fibrosis by reprogramming macrophage function through modulating the intracellular function of PKM2. Gut. 71:2539–2550. 2022. View Article : Google Scholar : PubMed/NCBI | |
Tu W, Ye J and Wang ZJ: Embryonic liver fordin is involved in glucose glycolysis of hepatic stellate cell by regulating PI3K/Akt signaling. World J Gastroenterol. 22:85192016. View Article : Google Scholar : PubMed/NCBI | |
Zhou MY, Cheng ML, Huang T, Hu RH, Zou GL, Li H, Zhang BF, Zhu JJ, Liu YM, Liu Y and Zhao XK: Transforming growth factor beta-1 upregulates glucose transporter 1 and glycolysis through canonical and noncanonical pathways in hepatic stellate cells. World J Gastroenterol. 27:6908–6926. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Chen L, Kong D, Zhang X, Xia S, Liang B, Li Y, Zhou Y, Zhang Z, Shao J, et al: Canonical Wnt signaling promotes HSC glycolysis and liver fibrosis through an LDH-A/HIF-1α transcriptional complex. Hepatology. 79:606–623. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wu X, Shen Y, Meng Y, Chen J, Zhang Y, Zeng S and Xu H: Suv39h1 contributes to activation of hepatic stellate cells in non-alcoholic fatty liver disease by enabling anaerobic glycolysis. Life Sci. 341:1224982024. View Article : Google Scholar : PubMed/NCBI | |
Huang T, Li YQ, Zhou MY, Hu RH, Zou GL, Li JC, Feng S, Liu YM, Xin CQ and Zhao XK: Focal adhesion kinase-related non-kinase ameliorates liver fibrosis by inhibiting aerobic glycolysis via the FAK/Ras/c-myc/ENO1 pathway. World J Gastroenterol. 28:123–139. 2022. View Article : Google Scholar : PubMed/NCBI | |
Xu L, Yang TY, Zhou YW, Wu MF, Shen J, Cheng JL, Liu QX, Cao SY, Wang JQ and Zhang L: Bmal1 inhibits phenotypic transformation of hepatic stellate cells in liver fibrosis via IDH1/α-KG-mediated glycolysis. Acta Pharmacol Sin. 43:316–329. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wan L, Xia T, Du Y, Liu J, Xie Y, Zhang Y, Guan F, Wu J, Wang X and Shi C: Exosomes from activated hepatic stellate cells contain GLUT1 and PKM2: A role for exosomes in metabolic switch of liver nonparenchymal cells. FASEB J. 33:8530–8542. 2019. View Article : Google Scholar : PubMed/NCBI | |
Khanal S, Liu Y, Bamidele AO, Wixom AQ, Washington AM, Jalan-Sakrikar N, Cooper SA, Vuckovic I, Zhang S, Zhong J, et al: Glycolysis in hepatic stellate cells coordinates fibrogenic extracellular vesicle release spatially to amplify liver fibrosis. Sci Adv. 10:eadn52282024. View Article : Google Scholar : PubMed/NCBI | |
Lian N, Jin H, Zhang F, Wu L, Shao J, Lu Y and Zheng S: Curcumin inhibits aerobic glycolysis in hepatic stellate cells associated with activation of adenosine monophosphate-activated protein kinase. IUBMB Life. 68:589–596. 2016. View Article : Google Scholar : PubMed/NCBI | |
Lian N, Jiang Y, Zhang F, Jin H, Lu C, Wu X, Lu Y and Zheng S: Curcumin regulates cell fate and metabolism by inhibiting hedgehog signaling in hepatic stellate cells. Lab Invest. 95:790–803. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wang F, Jia Y, Li M, Wang L, Shao J, Guo Q, Tan S, Ding H, Chen A, Zhang F and Zheng S: Blockade of glycolysis-dependent contraction by oroxylin a via inhibition of lactate dehydrogenase-a in hepatic stellate cells. Cell Commun Signal. 17:112019. View Article : Google Scholar : PubMed/NCBI | |
Ban D, Hua S, Zhang W, Shen C, Miao X and Liu W: Costunolide reduces glycolysis-associated activation of hepatic stellate cells via inhibition of hexokinase-2. Cell Mol Biol Lett. 24:522019. View Article : Google Scholar : PubMed/NCBI | |
Gao W, Sun J, Wang F, Lu Y, Wen C, Bian Q and Wu H: Deoxyelephantopin suppresses hepatic stellate cells activation associated with inhibition of aerobic glycolysis via hedgehog pathway. Biochem Biophys Res Commun. 516:1222–1228. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bae M, Lee Y, Pham TX, Hu S, Park YK and Lee JY: Astaxanthin inhibits the reduction of glycolysis during the activation of hepatic stellate cells. Life Sci. 256:1179262020. View Article : Google Scholar : PubMed/NCBI | |
Bae M, Kim MB and Lee JY: Fucoxanthin attenuates the reprogramming of energy metabolism during the activation of hepatic stellate cells. Nutrients. 14:19022022. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Zhou Y, Xia S, Chen L, Yang T, Zhao D, Zhang Z, Shao J, Xu X, Zhang F and Zheng S: Blockade of KLF5/LDH-A feedback loop contributes to Curcumol inhibition of sinusoidal endothelial cell glycolysis and mitigation of liver fibrosis. Phytomedicine. 114:1547592023. View Article : Google Scholar : PubMed/NCBI | |
Le CT, Nguyen G, Park SY, Dong HN, Cho YK, Lee JH, Im SS, Choi DH and Cho EH: Phloretin ameliorates succinate-induced liver fibrosis by regulating hepatic stellate cells. Endocrinol Metab (Seoul). 38:395–405. 2023. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Wang Y, Wan J, Dou X, Zhang C, Sun M and Ye F: Quercetin alleviates liver fibrosis via regulating glycolysis of liver sinusoidal endothelial cells and neutrophil infiltration. Biomol Biomed. 24:1806–1815. 2024. View Article : Google Scholar : PubMed/NCBI | |
Rho H, Terry AR, Chronis C and Hay N: Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis. Cell Metab. 35:1406–1423.e8. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li J, Chen X, Song S, Jiang W, Geng T, Wang T, Xu Y, Zhu Y, Lu J, Xia Y and Wang R: Hexokinase 2-mediated metabolic stress and inflammation burden of liver macrophages via histone lactylation in MASLD. Cell Rep. 44:1154652025. View Article : Google Scholar : PubMed/NCBI | |
Zhou Y, Yan J, Huang H, Liu L, Ren L, Hu J, Jiang X, Zheng Y, Xu L, Zhong F and Li X: The m6A reader IGF2BP2 regulates glycolytic metabolism and mediates histone lactylation to enhance hepatic stellate cell activation and liver fibrosis. Cell Death Dis. 15:1892024. View Article : Google Scholar : PubMed/NCBI | |
Wu S, Li J and Zhan Y: H3K18 lactylation accelerates liver fibrosis progression through facilitating SOX9 transcription. Exp Cell Res. 440:1141352024. View Article : Google Scholar : PubMed/NCBI | |
Tang F, Xiao D, Li X and Qiao L: The roles of lactate and the interplay with m6A modification in diseases. Cell Biol Toxicol. 40:1072024. View Article : Google Scholar : PubMed/NCBI | |
Chen W, Wang P, Xie Y, Xie D, Wang H, Bu N, Lin J, Wu M, Xia H, Cheng C, et al: Histone lactylation-augmented IRF4 is implicated in arsenite-induced liver fibrosis via modulating Th17 cell differentiation. Chem Biol Interact. 414:1115072025. View Article : Google Scholar : PubMed/NCBI | |
Kalantar-Zadeh K, Jafar TH, Nitsch D, Neuen BL and Perkovic V: Chronic kidney disease. Lancet. 398:786–802. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Zhang J, Feng D, Zhou H, Gui Z, Zheng M, Hang Z, Wang Z, Wang Z, Gu M and Tan R: IRF1/ZNF350/GPX4-mediated ferroptosis of renal tubular epithelial cells promote chronic renal allograft interstitial fibrosis. Free Radic Biol Med. 193:579–594. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li J, Zou Y, Kantapan J, Su H, Wang L and Dechsupa N: TGF-β/Smad signaling in chronic kidney disease: Exploring post-translational regulatory perspectives (review). Mol Med Rep. 30:1432024. View Article : Google Scholar : PubMed/NCBI | |
Ding H, Jiang L, Xu J, Bai F, Zhou Y, Yuan Q, Luo J, Zen K and Yang J: Inhibiting aerobic glycolysis suppresses renal interstitial fibroblast activation and renal fibrosis. Am J Physiol Renal Physiol. 313:F561–F575. 2017. View Article : Google Scholar : PubMed/NCBI | |
Yin XN, Wang J, Cui LF and Fan WX: Enhanced glycolysis in the process of renal fibrosis aggravated the development of chronic kidney disease. Eur Rev Med Pharmacol Sci. 22:4243–4251. 2018.PubMed/NCBI | |
Wang C, Li Y, Yan S, Wang H, Shao X, Xiao M, Yang B, Qin G, Kong R, Chen R and Zhang N: Interactome analysis reveals that lncRNA HULC promotes aerobic glycolysis through LDHA and PKM2. Nat Commun. 11:31622020. View Article : Google Scholar : PubMed/NCBI | |
Ashizawa K, Willingham MC, Liang CM and Cheng SY: In vivo regulation of monomer-tetramer conversion. J Biol Cem. 266:16842–16846. 1991.PubMed/NCBI | |
Liu H, Takagaki Y, Kumagai A, Kanasaki K and Koya D: The PKM2 activator TEPP-46 suppresses kidney fibrosis via inhibition of the EMT program and aberrant glycolysis associated with suppression of HIF-1α accumulation. J Diabetes Investig. 12:697–709. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Deng X, Liu Y, Liu Y, Sun L and Chen F: PKM2, function and expression and regulation. Cell Biosci. 9:522019. View Article : Google Scholar : PubMed/NCBI | |
Wang P, Sun C, Zhu T and Xu Y: Structural insight into mechanisms for dynamic regulation of PKM2. Protein Cell. 6:275–287. 2015. View Article : Google Scholar : PubMed/NCBI | |
Wei Q, Su J, Dong G, Zhang M, Huo Y and Dong Z: Glycolysis inhibitors suppress renal interstitial fibrosis via divergent effects on fibroblasts and tubular cells. Am J Physiol Renal Physiol. 316:F1162–F1172. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yu H, Zhu J, Chang L, Liang C, Li X and Wang W: 3-Bromopyruvate decreased kidney fibrosis and fibroblast activation by suppressing aerobic glycolysis in unilateral ureteral obstruction mice model. Life Sci. 272:1192062021. View Article : Google Scholar : PubMed/NCBI | |
Lee M, Harley G, Katerelos M, Gleich K, Sullivan MA, Laskowski A, Coughlan M, Fraser SA, Mount PF and Power DA: Mutation of regulatory phosphorylation sites in PFKFB2 worsens renal fibrosis. Sci Rep. 10:145312020. View Article : Google Scholar : PubMed/NCBI | |
Yang Q, Huo E, Cai Y, Zhang Z, Dong C, Asara JM and Wei Q: PFKFB3-mediated glycolysis boosts fibroblast activation and subsequent kidney fibrosis. Cells. 12:20812023. View Article : Google Scholar : PubMed/NCBI | |
Yang Q, Huo E, Cai Y, Zhang Z, Dong C, Asara JM, Shi H and Wei Q: Myeloid PFKFB3-mediated glycolysis promotes kidney fibrosis. Front Immunol. 14:12594342023. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Li H, Jiang S, Fu D, Lu X, Lu M, Li Y, Luo D, Wu K, Xu Y, et al: The glycolytic enzyme PFKFB3 drives kidney fibrosis through promoting histone lactylation-mediated NF-κB family activation. Kidney Int. 106:226–240. 2024. View Article : Google Scholar : PubMed/NCBI | |
Srivastava SP, Li J, Kitada M, Fujita H, Yamada Y, Goodwin JE, Kanasaki K and Koya D: SIRT3 deficiency leads to induction of abnormal glycolysis in diabetic kidney with fibrosis. Cell Death Dis. 9:9972018. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Wen P, Luo J, Ding H, Cao H, He W, Zen K, Zhou Y, Yang J and Jiang L: Sirtuin 3 regulates mitochondrial protein acetylation and metabolism in tubular epithelial cells during renal fibrosis. Cell Death Dis. 12:8472021. View Article : Google Scholar : PubMed/NCBI | |
Tang S, Huang T, Jing H, Huang Z, Chen H, Fan Y, Zhong J and Zhou J: Aldehyde dehydrogenase-2 acts as a potential genetic target for renal fibrosis. Life Sci. 239:1170152019. View Article : Google Scholar : PubMed/NCBI | |
Li SY, Tsai MT, Kuo YM, Yang HM, Tong ZJ, Cheng HW, Lin CC and Wang HT: Aldehyde dehydrogenase 2 preserves kidney function by countering acrolein-induced metabolic and mitochondrial dysfunction. JCI Insight. 9:e1798712024. View Article : Google Scholar : PubMed/NCBI | |
Cao H, Luo J, Zhang Y, Mao X, Wen P, Ding H, Xu J, Sun Q, He W, Dai C, et al: Tuberous sclerosis 1 (Tsc1) mediated mTORC1 activation promotes glycolysis in tubular epithelial cells in kidney fibrosis. Kidney Int. 98:686–698. 2020. View Article : Google Scholar : PubMed/NCBI | |
Li L, Galichon P, Xiao X, Figueroa-Ramirez AC, Tamayo D, Lee JJ, Kalocsay M, Gonzalez-Sanchez D, Chancay MS, McCracken KW, et al: Orphan nuclear receptor COUP-TFII enhances myofibroblast glycolysis leading to kidney fibrosis. EMBO Rep. 22:e511692021. View Article : Google Scholar : PubMed/NCBI | |
Cui X, Shi E, Li J, Li Y, Qiao Z, Wang Z, Liu M, Tang W, Sun Y, Zhang Y, et al: GPR87 promotes renal tubulointerstitial fibrosis by accelerating glycolysis and mitochondrial injury. Free Radic Biol Med. 189:58–70. 2022. View Article : Google Scholar : PubMed/NCBI | |
Li X, Ma TK, Wang M, Zhang XD, Liu TY, Liu Y, Huang ZH, Zhu YH, Zhang S, Yin L, et al: YY1-induced upregulation of LncRNA-ARAP1-AS2 and ARAP1 promotes diabetic kidney fibrosis via aberrant glycolysis associated with EGFR/PKM2/HIF-1α pathway. Front Pharmacol. 14:10693482023. View Article : Google Scholar : PubMed/NCBI | |
Yang S, Wu H, Li Y, Li L, Xiang J, Kang L, Yang G and Liang Z: Inhibition of PFKP in renal tubular epithelial cell restrains TGF-β induced glycolysis and renal fibrosis. Cell Death Dis. 14:8162023. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Tian M, Zhang M, Li C, Wang X, Long Y, Wang Y, Hu J, Chen C, Chen X, et al: Forkhead box Protein K1 promotes chronic kidney disease by driving glycolysis in tubular epithelial cells. Adv Sci. 24053252024. View Article : Google Scholar | |
Jiang A, Liu J, Wang Y and Zhang C: cGAS-STING signaling pathway promotes hypoxia-induced renal fibrosis by regulating PFKFB3-mediated glycolysis. Free Radic Biol Med. 208:516–529. 2023. View Article : Google Scholar : PubMed/NCBI | |
Hu D, Wang L, Zhang Y, Liu X, Lu Z and Li H: Sanqi oral solution ameliorates renal fibrosis by suppressing fibroblast activation via HIF-1α/PKM2/glycolysis pathway in chronic kidney disease. J Ethnopharmacol. 335:1186792024. View Article : Google Scholar : PubMed/NCBI | |
Cai T, Ke Q, Fang Y, Wen P, Chen H, Yuan Q, Luo J, Zhang Y, Sun Q, Lv Y, et al: Sodium-glucose cotransporter 2 inhibition suppresses HIF-1α-mediated metabolic switch from lipid oxidation to glycolysis in kidney tubule cells of diabetic mice. Cell Death Dis. 11:3902020. View Article : Google Scholar : PubMed/NCBI | |
Huang X, Gao L, Deng R, Peng Y, Wu S, Lu J and Liu X: Huangqi-Danshen decoction reshapes renal glucose metabolism profiles that delays chronic kidney disease progression. Biomed Pharmacother. 164:1149892023. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Feng X, Ye C, Wang C and Wang M: Shen Shuai II Recipe inhibits hypoxia-induced glycolysis by preserving mitochondrial dynamics to attenuate kidney fibrosis. J Ethnopharmacol. 308:1162712023. View Article : Google Scholar : PubMed/NCBI | |
Xie N, Tan Z, Banerjee S, Cui H, Ge J, Liu RM, Bernard K, Thannickal VJ and Liu G: Glycolytic reprogramming in myofibroblast differentiation and lung fibrosis. Am J Respir Crit Care Med. 192:1462–1474. 2015. View Article : Google Scholar : PubMed/NCBI | |
Kang YP, Lee SB, Lee J, Kim HM, Hong JY, Lee WJ, Choi CW, Shin HK, Kim DJ, Koh ES, et al: Metabolic profiling regarding pathogenesis of idiopathic pulmonary fibrosis. J Proteome Res. 15:1717–1724. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhao YD, Yin L, Archer S, Lu C, Zhao G, Yao Y, Wu L, Hsin M, Waddell TK, Keshavjee S, et al: Metabolic heterogeneity of idiopathic pulmonary fibrosis: A metabolomic study. BMJ Open Respir Res. 4:e0001832017. View Article : Google Scholar : PubMed/NCBI | |
Cho SJ, Moon JS, Lee CM, Choi AMK and Stout-Delgado HW: Glucose Transporter 1-Dependent glycolysis is increased during Aging-Related lung fibrosis, and phloretin inhibits lung fibrosis. Am J Respir Cell Mol Biol. 56:521–531. 2017. View Article : Google Scholar : PubMed/NCBI | |
Kim SH, Jin JA, So HJ, Lee SH, Kang TW, Lee JU, Choi DE, Jeong JY, Chang YK, Choi H, et al: Urine-derived stem Cell-secreted klotho plays a crucial role in the HK-2 fibrosis model by inhibiting the TGF-β signaling pathway. Int J Mol Sci. 23:50122022. View Article : Google Scholar : PubMed/NCBI | |
Yin X, Choudhury M, Kang JH, Schaefbauer KJ, Jung MY, Andrianifahanana M, Hernandez DM and Leof EB: Hexokinase 2 couples glycolysis with the profibrotic actions of TGF-β. Sci Signal. 12:eaax40672019. View Article : Google Scholar : PubMed/NCBI | |
Hu X, Xu Q, Wan H, Hu Y, Xing S, Yang H, Gao Y and He Z: PI3K-Akt-mTOR/PFKFB3 pathway mediated lung fibroblast aerobic glycolysis and collagen synthesis in lipopolysaccharide-induced pulmonary fibrosis. Lab Invest. 100:801–811. 2020. View Article : Google Scholar : PubMed/NCBI | |
Tang CJ, Xu J, Ye HY and Wang XB: Metformin prevents PFKFB3-related aerobic glycolysis from enhancing collagen synthesis in lung fibroblasts by regulating AMPK/mTOR pathway. Exp Ther Med. 21:5812021. View Article : Google Scholar : PubMed/NCBI | |
Chen W, Zhang J, Zhong W, Liu Y, Lu Y, Zeng Z, Huang H, Wan X, Meng X, Zou F, et al: Anlotinib Inhibits PFKFB3-Driven glycolysis in myofibroblasts to reverse pulmonary fibrosis. Front Pharmacol. 12:7448262021. View Article : Google Scholar : PubMed/NCBI | |
O'Leary EM, Tian Y, Nigdelioglu R, Witt LJ, Cetin-Atalay R, Meliton AY, Woods PS, Kimmig LM, Sun KA, Gökalp GA, et al: TGF-β promotes metabolic reprogramming in lung fibroblasts via mTORC1-dependent ATF4 activation. Am J Respir Cell Mol Biol. 63:601–612. 2020. View Article : Google Scholar : PubMed/NCBI | |
Xu J, Li J, Yu Z, Rao H, Wang S and Lan H: HMGB1 promotes HLF-1 proliferation and ECM production through activating HIF1-α-regulated aerobic glycolysis. Pulm Pharmacol Ther. 45:136–141. 2017. View Article : Google Scholar : PubMed/NCBI | |
Goodwin J, Choi H, Hsieh M, Neugent ML, Ahn JM, Hayenga HN, Singh PK, Shackelford DB, Lee IK, Shulaev V, et al: Targeting Hypoxia-Inducible Factor-1α/Pyruvate Dehydrogenase Kinase 1 Axis by dichloroacetate suppresses bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 58:216–231. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sun Z, Ji Z, He W, Duan R, Qu J and Yu G: Lactate accumulation induced by Akt2-PDK1 signaling promotes pulmonary fibrosis. FASEB J. 38:e234262024. View Article : Google Scholar : PubMed/NCBI | |
Schruf E, Schroeder V, Kuttruff CA, Weigle S, Krell M, Benz M, Bretschneider T, Holweg A, Schuler M, Frick M, et al: Human lung fibroblast-to-myofibroblast transformation is not driven by an LDH5-dependent metabolic shift towards aerobic glycolysis. Respir Res. 20:872019. View Article : Google Scholar : PubMed/NCBI | |
Mei S, Xu Q, Hu Y, Tang R, Feng J, Zhou Y, Xing S, Gao Y and He Z: Integrin β3-PKM2 pathway-mediated aerobic glycolysis contributes to mechanical ventilation-induced pulmonary fibrosis. Theranostics. 12:6057–6068. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Xu K, Wang N, Ding L, Zhao W, Wan R, Zhao W, Guo X, Pan X, Yang J, et al: Fenbendazole attenuates Bleomycin-induced pulmonary fibrosis in mice via suppression of Fibroblast-to-myofibroblast differentiation. Int J Mol Sci. 23:140882022. View Article : Google Scholar : PubMed/NCBI | |
Wang W, Zhang Y, Huang W, Yuan Y, Hong Q, Xie Z, Li L, Chen Y, Li X and Meng Y: Alamandine/MrgD axis prevents TGF-β1-mediated fibroblast activation via regulation of aerobic glycolysis and mitophagy. J Transl Med. 21:242023. View Article : Google Scholar : PubMed/NCBI | |
Lai X, Huang S, Lin Y, Qiu Y, Pu L, Lin S, Zeng Q, Huang W and Wang Z: DACT2 protects against pulmonary fibrosis via suppressing glycolysis in lung myofibroblasts. Int J Biol Macromol. 226:291–300. 2023. View Article : Google Scholar : PubMed/NCBI | |
Li J, Zeng G, Zhang Z, Wang Y, Shao M, Li C, Lu Z, Zhao Y, Zhang F and Ding W: Urban airborne PM2.5 induces pulmonary fibrosis through triggering glycolysis and subsequent modification of histone lactylation in macrophages. Ecotoxicol Environ Saf. 273:1161622024. View Article : Google Scholar : PubMed/NCBI | |
Feng J, Zhong H, Mei S, Tang R, Zhou Y, Xing S, Gao Y, Xu Q and He Z: LPS-induced monocarboxylate transporter-1 inhibition facilitates lactate accumulation triggering epithelial-mesenchymal transformation and pulmonary fibrosis. Cell Mol Life Sci. 81:2062024. View Article : Google Scholar : PubMed/NCBI | |
Gopu V, Fan L, Shetty RS, Nagaraja MR and Shetty S: Caveolin-1 scaffolding domain peptide regulates glucose metabolism in lung fibrosis. JCI Insight. 5:e1379692020. View Article : Google Scholar : PubMed/NCBI | |
Trivlidis J, Aloufi N, Al-Habeeb F, Nair P, Azuelos I, Eidelman DH and Baglole CJ: HuR drives lung fibroblast differentiation but not metabolic reprogramming in response to TGF-β and hypoxia. Respir Res. 22:3232021. View Article : Google Scholar : PubMed/NCBI | |
Xia Y, Wang Y, Xiong Q, He J, Wang H, Islam M, Zhou X, Kim A, Zhang H, Huang H and Tsung A: Neutrophil extracellular traps promote MASH fibrosis by metabolic reprogramming of HSC. Hepatology. 81:947–961. 2025. View Article : Google Scholar : PubMed/NCBI |