Open Access

Lactate and lactylation in the kidneys: Current advances and prospects (Review)

  • Authors:
    • Xu Li
    • Lan Hu
    • Qin Hu
    • Hua Jin
  • View Affiliations

  • Published online on: June 5, 2025     https://doi.org/10.3892/ijmm.2025.5562
  • Article Number: 121
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lactate, traditionally overlooked as a glycolytic byproduct, has recently been recognized for its significant biological roles. The normal kidney plays an essential role in maintaining systemic glucose and lactate homeostasis. Lactylation, as a new epigenetic modification, influences the initiation and progression of kidney diseases through the regulation of gene transcription and cellular metabolism. The present review summarizes current perspectives on the physiological functions of lactate and its renal metabolism, analyzes the roles of lactate and lactylation in acute kidney injury, diabetic nephropathy and chronic kidney disease, and proposes that targeted modulation of lactate metabolism may represent a promising therapeutic strategy for kidney disorders, thereby providing a foundation for future investigations.

Introduction

The kidney plays a vital role in numerous physiological processes, including the excretion of metabolic waste, the regulation of fluid balance and the secretion of endocrine hormones, which are all fundamental to maintaining internal homeostasis (1). In sugar metabolism, the kidneys contribute to acid-base equilibrium and energy supply by excreting and metabolizing lactate, thereby supporting glucose and lactate homeostasis (2). The involvement of lactate in kidney diseases includes intricate metabolic regulation, cellular injury and clinical monitoring. As a glycolytic product, lactate accumulates excessively under conditions of impaired clearance in renal dysfunction, potentially precipitating metabolic acidosis and directly impacting the function of renal tubular epithelial cells (3,4). During renal fibrosis, lactate accelerates fibrotic progression by inducing histone lactylation. For instance, in advanced glycation end product (AGE)-induced renal tubular epithelial cells, increased lactate levels enhance histone H3 lysine 14 lactylation (H3K14la) in diabetic nephropathy (DN), thereby promoting Krüppel-like factor 5 (KLF5) expression and driving the epithelial-mesenchymal transition (EMT) in DN (5). In chronic kidney disease (CKD), the expression of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) is positively correlated with the severity of renal fibrosis. PFKFB3-mediated glycolytic reprogramming in renal tubules generates lactate, which enhances H4K12la, facilitating gene transcription and activation of nuclear factor-κB (NF-κB), thus subsequently promoting inflammatory responses and exacerbating renal fibrosis (6). Furthermore, in acute kidney injury (AKI), DN and CKD, lactate concentrations and the activity or expression of glycolytic enzymes may exhibit alterations (4,7). Such metabolic shifts establish lactate-associated enzymes and lactate kinetics as emerging biomarkers for disease progression assessment. Consequently, lactate is not only a significant participant in renal metabolism but also a potential pathological driver in kidney disease evolution. Dynamic lactate monitoring offers valuable diagnostic and therapeutic insights, and targeting lactate metabolism may represent an innovative approach to renal protection.

Lactate, historically regarded as a negligible glycolytic byproduct with limited physiological relevance (8), has now emerged as a molecule of significant biological importance. Beyond its role as a ubiquitous metabolite, lactate functions as a substrate for epigenetic modifications, regulating gene transcription and protein function through lactylation of histone and non-histone proteins (9,10). This discovery illustrates the capacity of metabolites to regulate gene expression via post-translational modifications. Lactate, acting as a metabolic intermediate and signaling molecule, regulates diverse physiological and pathological processes, including osteoblast differentiation, inflammatory signal transduction, angiogenesis and myogenesis, among others (11-14). Notably, targeting the roles of lactate offers a promising therapeutic strategy for addressing chronic diseases. An expanding body of research highlights the relevance of lactate in various cellular functions, with particular attention on its emerging role in kidney disease.

The present review offers an in-depth evaluation of lactate and its associated metabolic pathways, providing a foundation to comprehending its role in renal physiology. Advances in delineating the involvement of lactate and lactylation in kidney disorders, specifically AKI, DN and CKD, are thoroughly evaluated. Moreover, an analysis of therapeutic targets and strategies focused on lactate metabolism and lactylation is performed, presenting innovative perspectives for the prevention and treatment of renal diseases.

Lactate and lactate metabolism

Generation and clearance of lactate

Glycolysis constitutes the primary pathway for lactate generation in organisms (15) (Fig. 1). Under hypoxic conditions, such as those experienced during intense physical exertion or within certain pathological environments, the entry of pyruvate into the mitochondria for aerobic metabolism is hindered. As a result, glucose in the cytoplasm undergoes enzymatic reactions leading to pyruvate formation, which is subsequently reduced to lactate through lactate dehydrogenase-A (LDHA). Notably, the glycolysis pathway can generate lactate even under aerobic conditions. This oxygen concentration-independent process represents a distinct metabolic mode known as aerobic glycolysis (16). The regulation of glycolysis is mediated by various enzymes, including LDH, pyruvate kinase M2 (PKM2), glucose transporters (GLUT) and hexokinase 2 (HK2) (17-20).

Lactate exists in two isomeric forms: L-lactate and D-lactate. L-lactate, the dominant isomer, is integral to immune regulation, acid-base balance and signal transduction within various physiological contexts (21). In eukaryotic cells, L-lactate represents the chief glycolytic end-product, whereas D-lactate is present only in trace amounts (22). D-lactate arises predominantly from intestinal microbiota metabolism, notably by Lactobacillus and Escherichia coli. In humans, lactate metabolism primarily depends on L-lactate dehydrogenase, while D-lactate dehydrogenase activity remains limited, resulting in slower D-lactate clearance (21,23).

Excessive lactate accumulation correlates with a heightened risk of lactic acidosis (24). Metabolic homeostasis necessitates the efficient clearance of lactate from tissues and the bloodstream (25,26). The primary mechanism for lactate clearance is its oxidation to pyruvate by LDHB in the presence of adequate oxygen, followed by mitochondrial entry for participation in the tricarboxylic acid cycle, yielding CO2, water and energy (27,28). Elevated lactate concentrations are also redirected to the liver and kidneys, where lactate undergoes conversion to glucose via gluconeogenesis. Notably, renal gluconeogenesis contributes ~40% of total gluconeogenesis during fasting, a function markedly compromised in CKD, leading to systemic metabolic imbalances manifesting as hypoglycemia and hyperlactatemia (7). Additionally, lactate is enzymatically transformed into fatty acids, cholesterol, and ketone bodies, or eliminated through the excretion of urine and sweat (Fig. 2).

Biological roles of lactate shuttle and lactate in the kidneys

The lactate shuttle theory systematically characterizes the intercellular transfer of lactate, highlighting its function as both an oxidative and gluconeogenic precursor, as well as a signaling molecule (29,30). This mechanism regulates the exchange of energy substrates between active cells, such as skeletal muscle, and recipient cells, including the brain, heart, liver and kidney, a process governed by the concentration gradients generated through mitochondrial respiration within recipient cells (30).

Monocarboxylate transporters (MCTs) are essential to the lactate shuttle mechanism, mediating lactate transport. The MCT family comprises 14 distinct members, each characterized by specific transport properties and tissue-specific expression. Among them, MCT1 and MCT4 are central to lactate dynamics: MCT1 promotes lactate flux in accordance with cellular metabolic needs, while MCT4 predominantly handles lactate efflux (31-33). Acting synergistically, these transporters maintain lactate homeostasis, pH balance and intracellular metabolic stability.

In murine kidneys, MCTs mediate the transport of monocarboxylates between renal tubular cells and the circulatory system (34). MCT1, MCT2, MCT7 and MCT8 are specifically localized on the basolateral membranes of the epithelial cells lining the nephron, with MCT1 and MCT8 expressed in proximal tubule cells, and MCT7 and MCT2 in the thick ascending limb and distal tubule (35). Concurrently, LDHA is predominantly expressed in the proximal nephron, functioning as a lactate producer, whereas LDHB is primarily localized to the distal nephron, serving as a lactate consumer, with both exhibiting spatial and temporal shifts in response to ischemic injury (36). Collectively, this spatial distribution supports the hypothesis that lactate shuttles from the proximal to the distal nephron, acting either as an energy substrate or a signaling mediator.

In addition, lactate exerts essential biological functions within the kidney. Podocytes and their foot processes constitute critical components of the renal filtration barrier, governing glomerular permeability. Podocyte injury is widely recognized as a central pathological mechanism in various kidney diseases, particularly DN and primary tubular disorders (37,38). Emerging evidence indicates that podocytes utilize lactate as an energy substrate and possess intrinsic regulatory systems to maintain lactate homeostasis. Under glucose-deprived conditions, L-lactate exposure preserves cellular viability and sustains glycolytic flux, mitigating glycogen depletion and highlighting the indispensable role of lactate in supporting podocyte metabolism during metabolic stress (39). Additionally, lactate stimulation modulates mitochondrial dynamics and respiratory efficiency; exposure to lactate elevates the mitochondrial DNA-to-nuclear DNA ratio, thereby enhancing mitochondrial biogenesis and regulating LDH subtype expression and activity in podocytes (40).

In renal cells, lactate not only undergoes catalysis to pyruvate, serving as a key intermediary in energy metabolism, but also functions as a gluconeogenic precursor for endogenous glucose synthesis. Renal gluconeogenesis predominantly occurs in the renal cortex, particularly within the proximal tubules, where the key enzymes required for the gluconeogenic pathway are expressed (41,42). Renal and hepatic gluconeogenesis collectively regulate systemic glucose homeostasis, critically contributing to the regulation of blood glucose levels and whole-body energy metabolism (43).

In clinical contexts, renal insufficiency impairs the lactate clearance capacity of the kidney, resulting in lactate accumulation and subsequent metabolic acidosis (44). This elevation serves as an indirect marker of renal dysfunction, particularly after excluding confounding factors such as hypoxia and infection. Therefore, monitoring lactate levels can indirectly help to assess the compensatory status of kidney function in patients with kidney disease. In diabetic populations, urinary lactate levels can predict clinical outcomes such as doubling of serum creatinine or the development of kidney failure, and elevated urinary lactate levels may be a potential biomarker for the risk of kidney disease progression (4). Thus, the role of lactate in kidney function not only reflects the metabolic and excretory capacity of the kidney but is also related to acidosis and disease prognosis. In clinical practice, a comprehensive analysis combining lactate levels, kidney function indicators and etiology should be conducted to optimize intervention strategies for kidney diseases.

In conclusion, lactate constitutes an essential metabolic substrate sustaining the energetic demands of renal cells. Beyond its fundamental bioenergetic contribution, lactate operates as a central regulator within cellular metabolic and signaling networks, critically supporting physiological stability and systemic metabolic balance.

Discovery of lactylation and its regulation

Lactate metabolism, homeostasis and the lactate microenvironment significantly regulate cellular and biomolecular functions. Recent studies have identified lactate as an epigenetic modulator (11,45,46), capable of inducing post-translational modifications and influencing gene expression, thereby causing diverse biological outcomes. Zhang et al (45) demonstrated that lactate facilitates transcriptional regulation through lactylation, an epigenetic mechanism wherein lactate-derived modifications of histone lysine residues directly promote gene transcription within chromatin, marking the initial discovery of histone lysine lactylation. Consequently, lactylation establishes a direct nexus between cellular metabolic activity and epigenetic governance.

Following the identification of histone lactylation, the emergence of non-histone lactylation has highlighted its wider prospects beyond chromatin modification (47). Current evidence implicates protein lactylation in a range of renal pathologies, including AKI (48) and DN (49), along with a variety of other diseases. For instance, H3K18la has been associated with the progression of arsenite-induced idiopathic pulmonary fibrosis (50), whereas glutamine mitigates intervertebral disc degeneration by inhibiting adenosine-5′-monophosphate-activated protein kinase α (AMPKα) lactylation through suppression of glycolytic pathways (51). Moreover, lactylation exerts regulatory effects on critical physiological processes, such as autophagy, macrophage activation and osteoblast differentiation (13,52,53). Accordingly, lactylation emerges as a central mechanism within both renal pathophysiology and systemic disease contexts.

The regulation of lactylation is modulated by a network of enzymes and regulatory proteins. Emerging evidence reveals that, analogous to acetylation, lactylation is controlled by 'writer' proteins such as P300/CREB binding protein and 'eraser' proteins, including histone deacetylases (HDACs) (13,54,55), and that these proteins are integral to the dynamic modulation and reversibility of lactylation (Fig. 3). P300, a major writer protein, catalyzes lysine lactylation by transferring lactate groups onto lysine residues. The P300/ASF1A complex integrates metabolic reprogramming with epigenetic modulation via H3K18la during atherogenesis driven by endothelial-to-mesenchymal transition (56). Eraser proteins, such as HDACs, counteract lactylation by enzymatically removing lactate groups. Notably, the feedback loop between H3K9la and HDAC2 in endothelial cells modulates vascular endothelial growth factor-induced angiogenesis; glycolysis inhibition reduces H3K9la levels and consequently attenuates neovascularization (11).

The identification of protein lactylation introduces a new framework for linking nutrient metabolism to gene expression. Continued investigation into lactylation and its regulatory pathways is anticipated to enhance the understanding of cellular dynamics and disease mechanisms. Expanding research efforts may clarify the functional roles of this post-translational modification and reveal novel therapeutic targets for related pathologies. Future studies should delineate the interplay between lactylation, cellular metabolism and other post-translational modifications, and systematically define the regulatory architecture of lactylation in kidney disease initiation and progression. Such advances would substantially enrich the mechanistic insight into lactate metabolism in kidney disease.

Detection methods of lactylation

Elucidating the biological significance of lactylation necessitates the development and implementation of precise detection methodologies.

Mass spectrometry (MS) is currently one of the most commonly used techniques for detecting lactylation, leveraging high-resolution instruments to identify mass shifts in proteins or peptides and thereby accurately pinpointing lactylation sites on proteins (57). As demonstrated by Li et al (58) in an AKI model, MS was successfully employed to identify lactylation sites on aldehyde dehydrogenase 2 (ALDH2), followed by immunoprecipitation-MS (IP-MS) analysis to characterize the interaction between ALDH2 and prohibitin 2 (PHB2). The study established that ALDH2 lactylation exacerbates mitochondrial dysfunction in AKI by impairing PHB2-mediated mitophagy (58).

Specific antibodies targeting lactylated lysine residues constitute indispensable tools for detecting lactylation, enabling the application of western blotting and IP assays to assess lactylation expression in cellular and tissue contexts. In a hypoxia/reoxygenation cell model in the human proximal tubular HK-2 cell line, Zhou et al (59) employed western blotting to detect histone lactylation and performed chromatin IP (ChIP) to assess its association with hexokinase 2 (HK2). The results demonstrated significant enrichment of H3K18la at the HK2 promoter region, which upregulated HK2 expression and consequently exacerbated renal ischemia/reperfusion injury. In renal tubular epithelial cells, An et al (48) demonstrated through western blot analysis that lactate overproduction mediates lactylation of mitochondrial fission 1 protein (Fis1) lysine 20 (Fis1 K20la), which subsequently induces mitochondrial dysfunction and exacerbates sepsis-associated AKI (SA-AKI).

Techniques such as immunofluorescence (IF) and immunohistochemistry (IHC) further allow for the visualization of the spatial distribution of lactylation within cells and tissues, providing an intuitive localization tool for studying lactylation. Qiao et al (60) employed western blotting and IF to assess H3K18la expression in the renal tubular epithelial cells of SA-AKI mice, combined with Cleavage Under Targets and Tagmentation technology to identify H3K18la-regulated target genes. The study revealed significant enrichment of H3K18la at the promoter region of Ras homolog gene family member A (RhoA), demonstrating that H3K18la upregulation activates the RhoA/Rho-associated protein kinase/Ezrin signaling pathway, thereby promoting renal dysfunction (60).

ChIP sequencing (ChIP-seq), a high-throughput method for analyzing protein-DNA interactions (61), facilitates genome-wide mapping of lactylation sites. Integration of RNA-seq data provides a complementary strategy to elucidate transcriptional regulatory networks influenced by lactylation modifications (62). As demonstrated by Zhang et al (5) in AGE-induced renal tubular epithelial cells, IHC and IF analyses of pan-lysine lactylation (pan-Kla) and H3K14la, combined with integrated ChIP-seq and RNA-seq profiling, revealed that H3K14la promotes the EMT process through upregulation of downstream KLF5 expression (5).

However, research on lactylation modifications remains in its nascent stages, with methodologies undergoing continuous refinement. Integrating multiple analytical approaches can significantly improve the reliability of findings. As technological advancements progress, future detection methods for lactylation modifications are expected to achieve greater sensitivity and efficiency, providing robust support for elucidating their significant roles in biology.

Function of lactylation in renal cells

The kidney has unique anatomical and physiological characteristics, with various renal cell types displaying specialized metabolic adaptations tailored to their specific functions and microenvironments (63). Consequently, the functional of lactylation in renal cells is likely determined by cell type-specific metabolic properties and pathological status, particularly in injury repair, metabolic regulation and fibrosis progression.

In AKI, the lactylation of renal tubular epithelial cells modulates kidney functional by regulating cellular proliferation and reparative pathways. For instance, heat shock protein A12A augments the proliferative capacity of renal tubular epithelial cells by enhancing c-Myc lactylation, thereby promoting functional restoration following AKI (64). Furthermore, lactylation influences the progression of SA-AKI, wherein lactate modifies inflammatory metabolism in renal proximal tubular epithelial cells via lactylation at H3K18 and Ezrin-K263 residues, consequently affecting SA-AKI recovery (60). Excessive lactate accumulation in renal tubular epithelial cells drives Fis1 K20la. Elevated Fis1 K20la levels promote aberrant mitochondrial fission, leading to ATP depletion, mitochondrial reactive oxygen species (ROS) overproduction and mitochondrial apoptosis, ultimately intensifying SA-AKI pathology (48). Thus, lactylation may regulate renal tubular epithelial cell survival and functional restoration in AKI through epigenetic mechanisms.

In DN, lactate derived from renal tubular epithelial cells promotes KLF5 expression via H3K14la, thereby upregulating Vimentin and α-smooth muscle actin (α-SMA) expression and accelerating the EMT process (5). In the CKD model, lactate stimulates transforming growth factor (TGF)-β1 expression in mouse renal tubular epithelial cells via H3K18la, which subsequently activates the Smad3 pathway in macrophages, driving macrophage-myofibroblast transition and renal fibrosis (65). Furthermore, in DN mice, lactate accumulation promotes histone pan-lactylation in podocytes, which downregulates nephrin and zonula occludens-1 levels while upregulating collagen IV, fibronectin and α-SMA, ultimately inducing EMT in podocytes (66).

These findings demonstrate that during AKI, renal tubular epithelial cells are susceptible to lactylation-induced disturbances in energy metabolism and oxidative stress. By contrast, under DN or CKD conditions, tubular lactylation appears to promote renal fibrogenesis. In podocytes, lactylation primarily drives fibrotic progression. Current evidence reveals the complex regulatory roles of lactylation in renal cells. However, its effects in other renal cells, including interstitial fibroblasts, glomerular mesangial cells, immune cells and pericytes, remain largely unexplored. The functional heterogeneity of lactylation among different renal cell populations warrants further investigation.

Lactate metabolism and lactylation in kidney diseases

AKI

AKI, a common and severe renal disorder, involves a sudden decline in renal function, evidenced by decreased glomerular filtration rate (GFR) and/or diminished urine output, resulting in metabolic waste accumulation, acid-base disturbances and electrolyte imbalances. Integrating prognostic biomarkers with clinical risk factors remains essential for early evaluation and management of AKI (67).

Clinical evidence has revealed an association between AKI and impaired gluconeogenesis, along with diminished lactate clearance (68). Elevated lactate levels have emerged as reliable predictors for both the onset and prognosis of AKI (69,70). Gong et al (71) identified increased serum lactate as an independent risk factor for SA-AKI, where concentrations ≥2.75 mmol/l were linked to a 1.772-fold escalation in AKI risk. In the postoperative setting, particularly after cardiac surgery, serum lactate levels independently predict AKI development (72). Early dynamic monitoring of lactate trajectories also enables the prediction of continuous renal replacement therapy requirements following acute type A aortic dissection surgery (73). Furthermore, in conditions such as acute decompensated heart failure (74), ST-segment elevation myocardial infarction (75) and traumatic brain injury (76), lactate levels serve as key indicators for assessing AKI risk, thereby strengthening opportunities for early diagnosis.

Clinical studies have highlighted the potential of lactate levels as biomarkers in AKI, prompting further exploration of the regulatory mechanisms of lactate in this condition. In a suppurative AKI model induced by cecal ligation and puncture in C57/B6 mice, lactate-mediated activation of the programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway triggered lymphocyte apoptosis and subsequent immunosuppression (Fig. 4), indicating that blockade of lactate receptors or inhibition of the PD-1/PD-L1 axis may offer innovative therapeutic strategies for septic AKI (77). Sepsis-induced aerobic glycolysis, characterized by increased lactate production and upregulation of glycolysis-associated genes in renal tissues, further implicates lactate in AKI pathology. In lipopolysaccharide-stimulated HK-2 cells, lactate treatment suppressed sirtuin 3 (SIRT3) and phosphorylated-AMP-activated protein kinase (p-AMPK) expression, decreased the microtubule-associated protein 1A/1B-light chain 3-II/microtubule-associated protein 1A/1B-light chain 3-I ratio and elevated p62 levels, thereby inhibiting autophagy, promoting apoptosis and intensifying sepsis-induced AKI (SI-AKI) progression (78) (Fig. 4). Intervention with the aerobic glycolysis inhibitor 2-deoxy-D-glucose attenuated glycolysis, with underlying mechanisms likely involving the restoration of autophagy via modulation of the lactate/SIRT3/AMPK signaling axis, thereby mitigating SI-AKI (78). Renal fibroblasts play a crucial role in the regulation, repair and recovery of renal tubular injury after AKI. The proliferation of fibroblasts follows the proliferation of renal tubular epithelial cells. Proliferating renal tubular epithelial cells preferentially utilize aerobic glycolysis as the primary metabolic pathway during AKI progression. In the later stages of AKI, lactate generated by injured tubular epithelial cells is absorbed by interstitial fibroblasts, promoting fibroblast activation and proliferation (79). Consequently, glycolysis-derived lactate from tubular epithelial cells modulates fibroblast activation and proliferation.

Figure 4

Mechanisms of lactate and lactylation in renal diseases. In a septic AKI mouse model, lactate activates the PD-1/PD-L1 pathway, inducing T lymphocyte apoptosis and contributing to immunosuppression during septic AKI. In LPS-stimulated HK-2 cells, lactate inhibits autophagy by downregulating SIRT3 and p-AMPK expression, reducing the LC3-II/I ratio and elevating p62 levels, thereby promoting HK-2 cell apoptosis and worsening SI-AKI. High acetylation and inactivation of PDHA1 in renal tubular epithelial cells drive lactate accumulation, mediate Fis1 K20la, impair mitochondrial function and aggravate SI-AKI. In human proximal tubular epithelial cells, increased lactylation at K182 of ACSF2 induces mitochondrial dysfunction, accelerates renal tubular injury and advances DN progression. Elevated lactate concentrations in renal tubular epithelial cells promote H3K14la, upregulating KLF5 expression and driving EMT in DN. In spontaneous hypertension, excessive lactate mediates renal fibrosis through activation of the TRPV4-TGFβ1-SMAD2/3-CTGF signaling cascade. ROS stress resulting from zinc deficiency also disrupts mitochondrial function, causing metabolic stress characterized by abnormal lactate metabolism and promoting the development of renal interstitial fibrosis. Moreover, lactate accumulation driven by PFKFB3 activation enhances H4K12la, augments NF-κB gene transcription and activation, intensifies inflammatory responses, and accelerates renal fibrosis. LPS, lipopolysaccharide; SIRT3, sirtuin 3; p-AMPK, phosphorylated-AMP-activated protein kinase; PDHA1, pyruvate dehydrogenase E1 component subunit α; Fis1 K20la, mitochondrial fission 1 protein lysine 20 lactylation; ACSF2, Acyl-CoA synthetase family member 2; K182la, lysine 182 lactylation; H3K14la, histone H3 lysine 14 lactylation; EMT, epithelial-mesenchymal transition; TRPV4, transient receptor potential vanilloid 4; TGF-β1, transforming growth factor β1; p-Smad2/3, phospho-Smad2/3; TRPV4, transient receptor potential cation channel subfamily V member 4; CTGF, connective tissue growth factor; ROS, reactive oxygen species; PFKFB3, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3; SI-AKI, sepsis-induced acute kidney injury; PD-1/PD-L1, programmed cell death protein 1/programmed death ligand 1; HK-2, hexokinase 2; LC3-II/I, microtubule-associated protein 1A/1B-light chain 3-II/microtubule-associated protein 1A/1B-light chain 3-I ratio; KLF5, Krüppel-like factor 5; diabetic neuropathy; CKD, chronic kidney disease. The figure was drawn using Figdraw (figdraw.com).

The discovery of lactylation has substantially advanced the understanding of its involvement in AKI. Elevated lactate levels are established as an independent risk factor for SA-AKI. Evidence suggests that H3K18la may serve as a biomarker for diagnosing and gauging the severity of septic shock (80). A recent investigation has reveal that increased H3K18la expression in SA-AKI contributes to the progression of renal insufficiency associated with this condition (60). Moreover, hyperacetylation and inactivation of pyruvate dehydrogenase E1 component subunit α in renal tubular epithelial cells promote excessive lactate production, subsequently inducing Fis1 lactylation and accelerating the deterioration of SI-AKI (48) (Fig. 4). These findings elucidate the epigenetic pathways implicated in AKI pathogenesis.

Although lactate is recognized as a biomarker for AKI prediction and prognosis assessment, the mechanisms underlying AKI progression and the specific contributions of lactate require further elucidation. While H3K18la involvement in SA-AKI has been characterized, the impact of lactylation in AKI resulting from other etiologies remains poorly defined. Future research should focus on delineating the roles and mechanistic pathways of lactate and lactylation across diverse AKI models.

DN

DN, a severe microvascular complication of diabetes characterized by proteinuria and progressive GFR decline, is a leading global contributor to CKD (81). Effective early detection and intervention depend on the development of robust predictive models and biomarkers. Accumulating evidence positions lactate as a promising non-invasive biomarker for early DN diagnosis (82). Jiang et al (83) proposed a DN risk prediction model incorporating serum lactate levels through a nomogram, offering a straightforward, economical and highly sensitive method for DN risk stratification.

Investigations into the role of LDH in DN have yielded significant insights. Tang et al (84) analyzed data from type 2 diabetes (T2DM) patients collected between 2009 and 2018 in the National Nutrition and Health Examination Survey database, utilizing restricted cubic spline plots to delineate a dose-response relationship between LDH levels and DN risk, thereby establishing LDH as a relevant biomarker for DN screening in T2DM populations. In parallel, Azushima et al (4) applied targeted metabolomics in a DN mouse model, revealing elevated renal lactate concentrations, impaired energy metabolism, and increased expression levels of LDHA and LDHB isoforms. Administration of angiotensin receptor blockers attenuated albuminuria, suppressed LDH expression, mitigated renal injury, restored metabolic homeostasis, normalized lactate concentrations and improved renal ATP content, implicating metabolic dysregulation and excessive lactate accumulation in the pathogenesis of DN.

Research into the mechanisms of lactate and lactylation in DN has demonstrated that enhanced lactylation at lysine 182 (K182) of ACSF2 in human proximal tubular epithelial cells induces mitochondrial dysfunction, thereby promoting renal tubular injury and accelerating DN progression (49) (Fig. 4). Moreover, EMT is widely recognized as a central contributor to DN development. In renal tubular epithelial cells exposed to AGEs, a metabolic shift from oxidative phosphorylation to glycolysis elevates renal lactate levels. Elevated lactate levels also increase H3K14la, subsequently upregulating KLF5 expression and further driving EMT in DN (5) (Fig. 4).

Advances in lactylation research have substantially expanded the understanding of the epigenetic regulation of DN. Modulation of the lactate-mediated H3K14la/KLF5 axis or adjustment of lysine lactylation levels offers a potential pathway for therapeutic innovation in DN. Although current evidence identifies lactate and LDH as candidate biomarkers for DN, further investigations are required to clarify the underlying mechanisms and enhance their clinical translation.

CKD

CKD constitutes a major global public health concern, characterized by chronic structural and functional renal impairments originating from diverse etiologies, including primary and secondary glomerulonephritis, tubular injury and renovascular pathologies (85,86).

Lactate metabolism holds clinical relevance in CKD across several dimensions, including metabolic dysregulation, prognostic evaluation and complication management. Impaired renal function, particularly tubular dysfunction, diminishes lactate clearance in patients with CKD. Evidence indicates that defective renal gluconeogenesis in CKD alters systemic metabolism, typified by reduced glucose and elevated lactate levels, increasing susceptibility to hyperlactatemia. This metabolic disturbance not only predisposes patients to metabolic acidosis but also correlates with adverse renal outcomes (7,87). Consequently, abnormal lactate metabolism is a manifestation of metabolic imbalance in patients with CKD. Monitoring blood lactate levels may help assess the status of lactate metabolism in patients with CKD and could potentially assist in evaluating the prognosis of the disease. However, its specific clinical application value in monitoring disease progression, managing complications and assessing prognosis in CKD still requires further evaluation.

The hallmark pathology of CKD is renal fibrosis, encompassing glomerulosclerosis, tubular atrophy and interstitial fibrosis. Dysregulated lactate metabolism has been implicated in fibrotic progression. In spontaneous hypertension, elevated lactate concentrations aggravate renal fibrosis via activation of the transient receptor potential cation channel subfamily V member 4-TGFβ1-SMAD2/3-connective tissue growth factor (CTGF) signaling cascade (88) (Fig. 4). Zinc, a critical micronutrient for mitigating oxidative stress and promoting tissue repair, is essential for maintaining systemic homeostasis. Markedly reduced plasma zinc concentrations have been reported in patients with CKD (89), where ROS stress resulting from zinc deficiency also disrupts mitochondrial function, causing metabolic stress characterized by abnormal lactate metabolism and promoting the development of renal interstitial fibrosis (90) (Fig. 4).

Enhanced renal glycolysis contributes to CKD progression (91), with PFKFB3 acting as a critical modulator of this metabolic shift (92). PFKFB3 expression is markedly upregulated in renal proximal tubular cells following ischemia-reperfusion injury in mice, displaying a positive correlation with the extent of renal fibrosis (6). Mechanistically, lactate accumulation driven by PFKFB3-mediated glycolytic reprogramming significantly elevates histone lactylation, particularly H4K12la, which is enriched at the promoters of NF-κB signaling genes, thereby initiating their transcription and intensifying inflammatory responses (6) (Fig. 4). Thus, targeting PFKFB3-mediated NF-κB signaling in tubular cells represents a potential therapeutic avenue for CKD. During renal fibrosis, cellular metabolic reprogramming occurs, with enhanced glycolysis constituting a major feature of disease progression (93,94). Suppressing glycolytic flux may, therefore, mitigate fibrosis advancement. However, glycolysis exerts divergent effects across different renal cells; for instance, dichloroacetic acid and shikonin inhibit fibroblast activation while eliciting distinct responses in renal epithelial cells (95), suggesting that cell type-specific metabolic modulation could open new therapeutic possibilities.

Recent investigations further implicate lactate metabolism in CKD progression, although the underlying mechanisms and clinical relevance require deeper exploration. Future research should prioritize identifying specific cell types affected by lactate dynamics, and delineating the gene networks and regulatory pathways modulated by lactate. Expanding this area of study could establish a theoretical framework for innovative therapeutic strategies and ultimately enhance clinical outcomes.

Prospects and limitations of lactate and its related biomarkers in nephropathy

Clinical application prospects of lactate-related biomarkers

The application prospects of lactate and its related biomarkers in samples of patients with kidney disease include the potential as diagnostic markers, prognostic assessment tools and therapeutic targets.

Evidence from a cohort of patients with T2DM demonstrated that urinary lactate levels maintained a statistically significant association with end-stage renal disease risk, even after adjustment for conventional risk factors such as estimated glomerular filtration rate and urine albumin-to-creatinine ratio, highlighting their value as predictive biomarkers for disease advancement (4). Notably, urinary lactate levels show a significant correlation with established biomarkers of tubular injury and epithelial stress, namely kidney injury molecule-1 and Dickkopf-3, suggesting its potential as a predictive biomarker for disease progression (4). These findings underscore the utility of urinary lactate as a potent tool for risk stratification in DN, where it is particularly adept at identifying patient populations who, despite presenting with seemingly acceptable conventional indicators, are actually at a high risk.

Beyond quantifying lactate concentrations, molecules involved in lactate metabolism have emerged as promising biomarkers. H3K14la, a lactate-mediated epigenetic modification, has been identified as a key participant in EMT progression in DN through activation of KLF5 expression (5). Theoretically, measuring H3K14la levels in renal tissues or urinary exosomes could serve as an indicator of renal fibrosis activity. However, standardized detection methodologies for this purpose remain unavailable.

The application of lactate-related biomarkers extends beyond diagnosis and prognosis, offering potential for therapeutic target identification. The elucidation of the H3K14la/KLF5 axis introduces new avenues for anti-fibrotic drug development, with interventions targeting this pathway representing a potential strategy to delay DN progression (5). In parallel, therapeutic approaches aimed at modulating lactate production or clearance, such as adjusting LDH activity or promoting lactate utilization, may offer new opportunities for kidney disease management. The refinement and implementation of these therapeutic strategies will inevitably rely on the guidance and monitoring of corresponding biomarkers.

Limitations of clinical application

Despite significant progress, current research and technological applications encounter multiple limitations that obstruct the clinical translation of biomarkers. Obstacles span the entire workflow, from sample collection and data analysis to mechanistic interpretation and clinical validation, demanding prompt and systematic resolution.

A primary technical barrier lies in the absence of standardized detection protocols. Studies utilize a wide range of lactate measurement techniques, including enzymatic electrode assays, gas chromatography-MS, and high-performance liquid chromatography, each differing in sensitivity, specificity and reproducibility, thereby complicating cross-study comparisons (96,97). Variations in sample types and analytical strategies further impede the definition of unified cutoff thresholds.

The invasive nature of sample collection constrains the clinical applicability of certain biomarkers. Although urinary lactate measurement offers a relatively non-invasive alternative, analyses of kidney tissue-specific modifications, such as H3K14la, continue to rely on renal biopsy specimens, limiting practicality for routine diagnostics. Even blood-based assays, while less invasive than tissue sampling, impose additional burdens due to the need for frequent collection, potentially compromising patient adherence. While microneedle sensors present a novel approach for minimally invasive monitoring, their long-term stability and reusability require further validation (98,99).

Regarding biomarker specificity, elevated lactate levels commonly arise under diverse pathophysiological conditions, including tissue hypoxia, hepatic dysfunction, systemic inflammatory responses and pharmacological influences (78,100). Such conditions may overlap in patients with renal disease, thereby limiting the utility of lactate quantification alone to accurately distinguish renal-specific injuries from systemic disturbances. Moreover, distinct kidney pathologies, such as DN and ischemic renal injury, may drive lactate metabolism alterations via different molecular mechanisms (63). However, the majority of current studies focus on specific disease types and lack systematic data for cross-disease comparisons.

Limitations in dynamic monitoring technologies constrain the clinical implementation of lactate-related biomarkers. Kidney disease progression often involves dynamic metabolic fluctuations that single-time-point measurements fail to capture. Although wearable devices theoretically support continuous monitoring, practical application remains hindered by challenges such as signal instability, skin irritation and acquisition efficiency (99,101).

The translational gap between mechanistic research and clinical application remains a major obstacle. Findings derived from basic studies often lack reproducibility in clinical cohorts. Additionally, current clinical investigations predominantly employ observational designs. Interventional trials are required to substantiate the clinical relevance of lactate-related biomarkers.

Research on lactate is evolving beyond its characterization as a metabolic byproduct toward its recognition as a multifunctional signaling molecule. Addressing existing challenges is essential for refining future research priorities, advancing detection methodologies and optimizing the clinical integration of current technologies. These efforts may offer novel perspectives for the early diagnosis and precision management of kidney diseases.

Management strategies of kidney diseases based on lactate metabolism

According to the present review, lactate has demonstrated predictive utility as a biomarker in kidney disease and contributes to disease progression; its production is regulated by critical glycolytic enzymes, including LDH, GLUT, HK2 and PKM2. Modulation of lactate metabolism through targeting these enzymes, altering its synthesis, transport and conversion, represents a potential therapeutic approach for the treatment of kidney diseases.

PKM2, a key isomer of the pyruvate kinase family, serves a fundamental role in glycolysis; its expression is markedly diminished in podocytes from patients with hypertensive nephropathy and DN (102). Urinary PKM2 has also emerged as a potential biomarker for predicting SA-AKI (103), highlighting PKM2 as an attractive therapeutic target for kidney diseases. TEPP-46, a small-molecule activator of PKM2, enhances glycolytic flux in healthy renal tissue (104). In diabetic mouse models, TEPP-46 restores tubular epithelial integrity by suppressing EMT and abnormal glycolysis, thereby attenuating diabetic renal fibrosis (105). Additionally, TEPP-46 restricts pericyte proliferation, migration and pericyte-myofibroblast transdifferentiation by limiting nuclear translocation of PKM2, offering a new approach to preventing the transition from AKI to CKD (106). Microcystin-RR (MC-RR), which demonstrates anti-pulmonary fibrosis properties (107), also exhibits renal protective effects. Using unilateral ureteral obstruction (UUO) mouse models and in vitro cell models, Ren et al (108) demonstrated that MC-RR directly bound to PKM2, modulated the PKM2-HIF-1α signaling axis, restored suppressed MMP-7 and MMP-13 expression, and reduced elevated MMP-9 levels in UUO renal tissue, thereby mitigating renal fibrosis. Furthermore, several Chinese herbal compounds exert nephroprotective effects through PKM2 targeting. For instance, modified Hu-lu-ba-wan ameliorates glomerular injury and podocyte apoptosis by maintaining PKM2-mediated mitochondrial homeostasis in DN (109). Similarly, Qian Yang Yu Yin granules improve hypertensive nephropathy by reprogramming metabolism through the HIF-1α/PKM2 positive feedback mechanism (110).

HK2, the principal rate-limiting enzyme of glycolysis, serves a central function in metabolic regulation. Evidence indicates that ceria nanoparticles attenuate metabolic disturbances in renal fibrosis models by diminishing ROS, enhancing mitochondrial ATP production, and suppressing HK1 and HK2 expression, thereby conferring renal protection (111). LDHA, responsible for catalyzing the conversion of pyruvate to lactate during glycolysis, is integral to maintaining cellular energy equilibrium. Curcumin, the primary curcuminoid in turmeric, mediates renoprotective effects by inhibiting aerobic glycolysis through the miR-489/LDHA axis, thereby alleviating glucose fluctuation-induced renal injury in the 293 cell model (112). Moreover, the glycolysis inhibitor 3-bromopyruvate restrains TGF-β1-induced fibroblast proliferation in a time- and dose-dependent manner, significantly downregulating the expression of aerobic glycolysis-related enzymes, such as HK2, LDHA and PKM2, while modulating the IL-1 receptor-associated kinase 4/MYC signaling pathway to mitigate renal fibrosis (113).

Modulating glucose entry into glycolysis represents a promising therapeutic strategy. The membrane protein GLUT is integral to the transport of glucose and related substrates across cell membranes. In HK-2 cells treated with high glucose, the expression level of GLUT-1 was upregulated (114), and in a streptozotocin-induced DN rat model, the expression level of GLUT-2 was downregulated (115). p-Coumaric acid nanoparticles have been shown to attenuate DN in rats by controlling hyperglycemia, suppressing inflammation and enhancing GLUT-2 mRNA expression in nephropathic models (115). Similarly, Huangqi decoction restores GLUT expression in diabetic kidneys, elevating GLUT4 levels while reducing GLUT1 expression in a dose-dependent manner (116). A bioflavonoid combination has further demonstrated renal function improvement in DN by regulating MMP-9/TIMP-1 expression, upregulating GLUT-4 and downregulating TGF-β (117). Regulation of glycolytic enzymes through these interventions may influence lactate production, transport and metabolism, offering therapeutic advantages in kidney disease management.

Clinical research into drugs targeting lactate metabolism or modulating lactylation mechanisms for kidney disease treatment remains an emerging and promising domain. Pharmacological or metabolic interventions designed to reduce lactate accumulation may potentially slow disease progression. Sodium-glucose cotransporter 2 (SGLT2) inhibitors, such as dapagliflozin, currently in clinical use, appear to indirectly decrease lactate levels by enhancing energy metabolism and exert renal protective effects in patients with DN (118). Levocarnitine mitigates oxidative stress and attenuates lactate accumulation in patients with kidney disease, particularly benefiting individuals with chronic renal failure and L-carnitine deficiency secondary to hemodialysis (119,120). However, most agents intended to regulate lactate metabolism or influence lactylation pathways remain at experimental stages, with clinical validation of their therapeutic potential still limited. Further research is warranted to clarify their mechanisms of action, assess safety profiles and establish therapeutic efficacy to support clinical translation in kidney disease management.

In clinical practice, dysregulation of lactate metabolism and lactate-mediated modifications in the kidney constitute a shared pathophysiological basis for multiple renal disorders. Effective monitoring requires an integrated evaluation of blood lactate concentrations, urinary lactate levels and biomarkers of lactate modifications in renal biopsy specimens. Management of patients with lactic acidosis emphasizes the aggressive treatment of underlying conditions, correction of hypoxic states and amelioration of metabolic perturbations. In the medication of certain populations, particular caution is warranted in patients with CKD to avoid nephrotoxins that may enhance lactate production, such as metformin (121), alongside monitoring of serum lactate and acid-base status. In the context of SA-AKI, early goal-directed therapy combined with infection source control can mitigate lactate accumulation. Pharmacological strategies targeting lactification currently remain under investigation. Future progress will depend on interdisciplinary efforts to integrate lactate metabolic regulation with epigenetic modification mechanisms, thereby enhancing their translational potential.

Targeting glycolytic enzymes to regulate lactate metabolism offers a promising therapeutic approach for kidney diseases but faces multiple substantive challenges. Low target specificity remains a major concern, as glycolytic enzymes such as PKM2 and HK2 are extensively involved in systemic metabolism, raising the risk of unintended metabolic disturbances. The pronounced heterogeneity among kidney diseases further complicates therapeutic outcomes, with the same target having variable effects across different pathological contexts. Insufficient drug delivery efficiency also limits therapeutic success, particularly for natural compounds such as curcumin, which exhibit poor renal targeting due to hepatic first-pass metabolism and systemic distribution. Additionally, inhibition of a single glycolytic enzyme may provoke compensatory metabolic pathways, introducing further complexity into therapeutic modulation. Clinical translation remains constrained by the reliance on animal and in vitro models, with minimal validation in human kidney tissues. Future strategies require the development of highly specific renal delivery technologies. Integrating multi-omics approaches to delineate the metabolic-immune-epigenetic networks, alongside the construction of dynamic biomarker systems through spatial metabolomics, will be instrumental in mapping the spatiotemporal heterogeneity of lactate metabolism throughout kidney disease progression. Furthermore, advancing personalized therapeutic strategies through clinical trials guided by dynamic biomarkers may shift the paradigm from merely correcting metabolic imbalances to reconstructing renal microenvironmental homeostasis.

Conclusion

Lactate and its metabolic pathways exert multifaceted roles in renal physiology and pathology, functioning not only as central mediators of energy metabolism but also through lactylation-driven epigenetic modifications that influence renal cell inflammation, fibrosis and reparative processes. Current research has preliminarily established the association between dysregulated lactate metabolism and kidney diseases, with interventions targeting lactate pathways exhibiting therapeutic potential in preclinical models. Nevertheless, prevailing conclusions largely derive from broad-spectrum metabolic regulation studies, lacking precise delineation of cell-type specificity, spatiotemporal dynamics and the molecular network underlying lactylation-mediated pathological mechanisms, which remain to be systematically elucidated.

Future research priorities should address the following areas: i) Development of precision intervention tools, engineering tissue-specific modulators of lactylation, such as LDHA inhibitors or lactylation enzyme regulators targeting renal tubular epithelial cells, to enable selective intervention within renal parenchymal cells while minimizing systemic metabolic perturbations. ii) Construction of dynamic models, establishing animal models capturing the temporal dynamics of lactylation alterations during kidney disease progression, including cell-specific lactate sensor transgenic mice to characterize metabolic heterogeneity across pathological stages. iii) Integration of interdisciplinary technologies, applying multi-omics approaches such as metabolomics and spatial transcriptomics to unravel the interaction networks linking the glycolysis-lactate axis with immune microenvironmental remodeling and fibrotic pathways, and identifying key regulatory nodes. Concurrently, clinical translation challenges must be addressed by developing non-invasive detection tools for lactate metabolism markers and validating the safety profiles of therapeutic strategies using models such as organoids, thereby providing a basis for personalized treatment. Comprehensive, multidimensional analyses of cell-specific mechanisms and dynamic lactate metabolic networks are anticipated to drive the evolution from biomarker discovery to targeted therapies in kidney disease management.

Availability of data and materials

Not applicable.

Authors' contributions

XL wrote the manuscript text. HJ conceived and designed the review. LH and QH edited and revised the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

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.

Acknowledgments

Not applicable.

Funding

This study was supported by the National Natural Science Foundation of China (grant no. 82274307) and the Research Funds of Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of Institute of Health and Medicine (grant no. 2023CXMMTCM018).

References

1 

Silva PHI and Mohebbi N: Kidney metabolism and acid-base control: Back to the basics. Pflugers Arch. 474:919–934. 2022. View Article : Google Scholar

2 

Chen Y, Fry BC and Layton AT: Modeling glucose metabolism and lactate production in the kidney. Math Biosci. 289:116–129. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Reddy AJ, Lam SW, Bauer SR and Guzman JA: Lactic acidosis: Clinical implications and management strategies. Clevel Clin J Med. 82:615–624. 2015. View Article : Google Scholar

4 

Azushima K, Kovalik JP, Yamaji T, Ching J, Chng TW, Guo J, Liu JJ, Nguyen M, Sakban RB, George SE, et al: Abnormal lactate metabolism is linked to albuminuria and kidney injury in diabetic nephropathy. Kidney Int. 104:1135–1149. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Zhang X, Chen J, Lin R, Huang Y, Wang Z, Xu S, Wang L, Chen F, Zhang J, Pan K and Yin Z: Lactate drives epithelial-mesenchymal transition in diabetic kidney disease via the H3K14la/KLF5 pathway. Redox Biol. 75:1032462024. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Verissimo T, Faivre A, Rinaldi A, Lindenmeyer M, Delitsikou V, Veyrat-Durebex C, Heckenmeyer C, Fernandez M, Berchtold L, Dalga D, et al: Decreased renal gluconeogenesis is a hallmark of chronic kidney disease. J Am Soc Nephrol. 33:810–827. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Rabinowitz JD and Enerbäck S: Lactate: The ugly duckling of energy metabolism. Nat Metab. 2:566–571. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Merkuri F, Rothstein M and Simoes-Costa M: Histone lactylation couples cellular metabolism with developmental gene regulatory networks. Nat Commun. 15:902024. View Article : Google Scholar : PubMed/NCBI

10 

Li J, Hou W, Zhao Q, Han W, Cui H, Xiao S, Zhu L, Qu J, Liu X, Cong W, et al: Lactate regulates major zygotic genome activation by H3K18 lactylation in mammals. Natl Sci Rev. 11:nwad2952024. View Article : Google Scholar : PubMed/NCBI

11 

Dai W, Wu G, Liu K, Chen Q, Tao J, Liu H and Shen M: Lactate promotes myogenesis via activating H3K9 lactylation-dependent up-regulation of Neu2 expression. J Cachexia Sarcopenia Muscle. 14:2851–2865. 2023. View Article : Google Scholar : PubMed/NCBI

12 

Fan W, Zeng S, Wang X, Wang G, Liao D, Li R, He S, Li W, Huang J, Li X, et al: A feedback loop driven by H3K9 lactylation and HDAC2 in endothelial cells regulates VEGF-induced angiogenesis. Genome Biol. 25:1652024. View Article : Google Scholar : PubMed/NCBI

13 

Minami E, Sasa K, Yamada A, Kawai R, Yoshida H, Nakano H, Maki K and Kamijo R: Lactate-induced histone lactylation by p300 promotes osteoblast differentiation. PLoS One. 18:e02936762023. View Article : Google Scholar : PubMed/NCBI

14 

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

15 

Kierans SJ and Taylor CT: Glycolysis: A multifaceted metabolic pathway and signaling hub. J Biol Chem. 300:1079062024. View Article : Google Scholar

16 

Luengo A, Li Z, Gui DY, Sullivan LB, Zagorulya M, Do BT, Ferreira R, Naamati A, Ali A, Lewis CA, et al: Increased demand for NAD+ relative to ATP drives aerobic glycolysis. Mol Cell. 81:691–707.e6. 2021. View Article : Google Scholar :

17 

Wang L, Pavlou S, Du X, Bhuckory M, Xu H and Chen M: Glucose transporter 1 critically controls microglial activation through facilitating glycolysis. Mol Neurodegener. 14:22019. View Article : Google Scholar : PubMed/NCBI

18 

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

19 

Nishioku T, Anzai R, Hiramatsu S, Terazono A, Nakao M and Moriyama M: Lactate dehydrogenase A inhibition prevents RANKL-induced osteoclastogenesis by reducing enhanced glycolysis. J Pharmacol Sci. 153:197–207. 2023. View Article : Google Scholar : PubMed/NCBI

20 

Kim E, Hwang Y, Kim H, Kim GU, Ryu YC, Yoon M and Choi KY: Pyruvate Kinase M2 accelerates cutaneous wound healing via glycolysis and Wnt/β-catenin signaling. Pharmaceutics. 15:20282023. View Article : Google Scholar

21 

Li J, Ma P, Liu Z and Xie J: L- and D-lactate: Unveiling their hidden functions in disease and health. Cell Commun Signal. 23:1342025. View Article : Google Scholar : PubMed/NCBI

22 

Heim CE, Bosch ME, Yamada KJ, Aldrich AL, Chaudhari SS, Klinkebiel D, Gries CM, Alqarzaee AA, Li Y, Thomas VC, et al: Lactate production by Staphylococcus aureus biofilm inhibits HDAC11 to reprogram the host immune response during persistent infection. Nat Microbiol. 5:1271–1284. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Monroe GR, van Eerde AM, Tessadori F, Duran KJ, Savelberg SMC, van Alfen JC, Terhal PA, van der Crabben SN, Lichtenbelt KD, Fuchs SA, et al: Identification of human D lactate dehydrogenase deficiency. Nat Commun. 10:14772019. View Article : Google Scholar : PubMed/NCBI

24 

Vernon C and LeTourneau JL: Lactic acidosis: Recognition, kinetics, and associated prognosis. Critical Care Clinics. 26:255–283. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Emhoff CAW and Messonnier LA: Concepts of lactate metabolic clearance rate and lactate clamp for metabolic inquiry: A Mini-review. Nutrients. 15:32132023. View Article : Google Scholar : PubMed/NCBI

26 

Huang T, Liang Z, Wang K, Miao X and Zheng L: Novel insights into athlete physical recovery concerning lactate metabolism, lactate clearance and fatigue monitoring: A comprehensive review. Front Physiol. 16:14597172025. View Article : Google Scholar : PubMed/NCBI

27 

Lin Y, Wang Y and Li P: Mutual regulation of lactate dehydrogenase and redox robustness. Front Physiol. 13:10384212022. View Article : Google Scholar : PubMed/NCBI

28 

Adeva M, González-Lucán M, Seco M and Donapetry C: Enzymes involved in l-lactate metabolism in humans. Mitochondrion. 13:615–629. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Wei T, Guo Y, Huang C, Sun M, Zhou B, Gao J and Shen W: Fibroblast-to-cardiomyocyte lactate shuttle modulates hypertensive cardiac remodelling. Cell Biosci. 13:1512023. View Article : Google Scholar : PubMed/NCBI

30 

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

31 

Zhang L, Xin C, Wang S, Zhuo S, Zhu J, Li Z, Liu Y, Yang L and Chen Y: Lactate transported by MCT1 plays an active role in promoting mitochondrial biogenesis and enhancing TCA flux in skeletal muscle. Sci Adv. 10:eadn45082024. View Article : Google Scholar : PubMed/NCBI

32 

Contreras-Baeza Y, Sandoval PY, Alarcón R, Galaz A, Cortés-Molina F, Alegría K, Baeza-Lehnert F, Arce-Molina R, Guequén A, Flores CA, et al: Monocarboxylate transporter 4 (MCT4) is a high affinity transporter capable of exporting lactate in high-lactate microenvironments. J Biol Chem. 294:20135–20147. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Kobayashi M, Narumi K, Furugen A and Iseki K: Transport function, regulation, and biology of human monocarboxylate transporter 1 (hMCT1) and 4 (hMCT4). Pharmacol Ther. 226:1078622021. View Article : Google Scholar : PubMed/NCBI

34 

Yanase H, Takebe K, Nio-Kobayashi J, Takahashi-Iwanaga H and Iwanaga T: Cellular expression of a sodium-dependent monocarboxylate transporter (Slc5a8) and the MCT family in the mouse kidney. Histochem Cell Biol. 130:957–966. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Becker HM, Mohebbi N, Perna A, Ganapathy V, Capasso G and Wagner CA: Localization of members of MCT monocarboxylate transporter family Slc16 in the kidney and regulation during metabolic acidosis. Am J Physiol Renal Physiol. 299:F141–F154. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Osis G, Traylor AM, Black LM, Spangler D, George JF, Zarjou A, Verlander JW and Agarwal A: Expression of lactate dehydrogenase A and B isoforms in the mouse kidney. Am J Physiol Renal Physiol. 320:F706–F718. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Feng Y, Sun Z, Fu J, Zhong F, Zhang W, Wei C, Chen A, Liu BC, He JC and Lee K: Podocyte-derived soluble RARRES1 drives kidney disease progression through direct podocyte and proximal tubular injury. Kidney Int. 106:50–66. 2024. View Article : Google Scholar : PubMed/NCBI

38 

Zhao Y, Fan S, Zhu H, Zhao Q, Fang Z, Xu D, Lin W, Lin L, Hu X, Wu G, et al: Podocyte OTUD5 alleviates diabetic kidney disease through deubiquitinating TAK1 and reducing podocyte inflammation and injury. Nat Commun. 15:54412024. View Article : Google Scholar : PubMed/NCBI

39 

Szrejder M, Typiak M, Pikul P, Audzeyenka I, Rachubik P, Rogacka D, Narajczyk M and Piwkowska A: Role of L-lactate as an energy substrate in primary rat podocytes under physiological and glucose deprivation conditions. Eur J Cell Biol. 102:1512982023. View Article : Google Scholar : PubMed/NCBI

40 

Audzeyenka I, Szrejder M, Rachubik P, Grochowalska K, Kulesza T, Rogacka D, Narajczyk M and Piwkowska A: Lactate regulates respiratory efficiency and mitochondrial dynamics in primary rat podocytes. Free Radic Biol Med. 220:312–323. 2024. View Article : Google Scholar : PubMed/NCBI

41 

Dalga D, Verissimo T and de Seigneux S: Gluconeogenesis in the kidney: In health and in chronic kidney disease. Clin Kidney J. 16:1249–1257. 2023. View Article : Google Scholar : PubMed/NCBI

42 

Nakamura M, Satoh N, Horita S and Nangaku M: Insulin-induced mTOR signaling and gluconeogenesis in renal proximal tubules: A mini-review of current evidence and therapeutic potential. Front Pharmacol. 13:10152042022. View Article : Google Scholar : PubMed/NCBI

43 

Hatano R, Lee E, Sato H, Kiuchi M, Hirahara K, Nakagawa Y, Shimano H, Nakayama T, Tanaka T and Miki T: Hepatic ketone body regulation of renal gluconeogenesis. Mol Metab. 84:1019342024. View Article : Google Scholar : PubMed/NCBI

44 

Zanza C, Facelli V, Romenskaya T, Bottinelli M, Caputo G, Piccioni A, Franceschi F, Saviano A, Ojetti V, Savioli G and Longhitano Y: Lactic acidosis related to pharmacotherapy and human diseases. Pharmaceuticals (Basel). 15:14962022. View Article : Google Scholar : PubMed/NCBI

45 

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

46 

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 :

47 

Wan N, Wang N, Yu S, Zhang H, Tang S, Wang D, Lu W, Li H, Delafield DG, Kong Y, et al: Cyclic immonium ion of lactyllysine reveals widespread lactylation in the human proteome. Nat Methods. 19:854–864. 2022. View Article : Google Scholar : PubMed/NCBI

48 

An S, Yao Y, Hu H, Wu J, Li J, Li L, Wu J, Sun M, Deng Z, Zhang Y, et al: PDHA1 hyperacetylation-mediated lactate overproduction promotes sepsis-induced acute kidney injury via Fis1 lactylation. Cell Death Dis. 14:4572023. View Article : Google Scholar : PubMed/NCBI

49 

Chen J, Feng Q, Qiao Y, Pan S, Liang L, Liu Y, Zhang X, Liu D and Liu Z and Liu Z: ACSF2 and lysine lactylation contribute to renal tubule injury in diabetes. Diabetologia. 67:1429–1443. 2024. View Article : Google Scholar : PubMed/NCBI

50 

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

51 

Zhang Y, Huang Z, Han W, Wu J, Li S, Qin T, Zhang C, Shi M, Han S, Gao B, et al: Glutamine suppresses senescence and promotes autophagy through glycolysis inhibition-mediated AMPKα lactylation in intervertebral disc degeneration. Commun Biol. 7:3252024. View Article : Google Scholar

52 

Sun W, Jia M, Feng Y and Cheng X: Lactate is a bridge linking glycolysis and autophagy through lactylation. Autophagy. 19:3240–3241. 2023. View Article : Google Scholar : PubMed/NCBI

53 

Wei Y, Guo H, Chen S and Tang XX: Regulation of macrophage activation by lactylation in lung disease. Front Immunol. 15:14277392024. View Article : Google Scholar : PubMed/NCBI

54 

Moreno-Yruela C, Zhang D, Wei W, Bæk M, Liu W, Gao J, Danková D, Nielsen AL, Bolding JE, Yang L, et al: Class I histone deacetylases (HDAC1-3) are histone lysine delactylases. Sci Adv. 8:eabi66962022. View Article : Google Scholar : PubMed/NCBI

55 

Kikuchi M, Morita S, Wakamori M, Sato S, Uchikubo-Kamo T, Suzuki T, Dohmae N, Shirouzu M and Umehara T: Epigenetic mechanisms to propagate histone acetylation by p300/CBP. Nat Commun. 14:41032023. View Article : Google Scholar : PubMed/NCBI

56 

Dong M, Zhang Y, Chen M, Tan Y, Min J, He X, Liu F, Gu J, Jiang H, Zheng L, et al: ASF1A-dependent P300-mediated histone H3 lysine 18 lactylation promotes atherosclerosis by regulating EndMT. Acta Pharm Sin B. 14:3027–3048. 2024. View Article : Google Scholar : PubMed/NCBI

57 

Wu X and Tao WA: Uncovering ubiquitous protein lactylation. Nat Methods. 19:793–794. 2022. View Article : Google Scholar : PubMed/NCBI

58 

Li J, Shi X, Xu J, Wang K, Hou F, Luan X and Chen L: Aldehyde dehydrogenase 2 lactylation aggravates mitochondrial dysfunction by disrupting PHB2 mediated mitophagy in acute kidney injury. Adv Sci (Weinh). 12:e24119432024. View Article : Google Scholar : PubMed/NCBI

59 

Zhou J, Zhang J, Xu F, Gao H, Wang L, Zhao Y and Li K: AST-120 alleviates renal ischemia-reperfusion injury by inhibiting HK2-mediated glycolysis. Mol Med. 30:1332024. View Article : Google Scholar : PubMed/NCBI

60 

Qiao J, Tan Y, Liu H, Yang B, Zhang Q, Liu Q, Sun W, Li Z, Wang Q, Feng W, et al: Histone H3K18 and ezrin lactylation promote renal dysfunction in Sepsis-associated acute kidney injury. Adv Sci (Weinh). 11:e23072162024. View Article : Google Scholar : PubMed/NCBI

61 

Kumar B, Navarro C, Yung PYK, Lyu J, Salazar Mantero A, Katsori AM, Schwämmle H, Martin M and Elsässer SJ: Multiplexed chromatin immunoprecipitation sequencing for quantitative study of histone modifications and chromatin factors. Nat Protoc. 20:779–809. 2025. View Article : Google Scholar

62 

Zhang L, Xue G, Liu J, Li Q and Wang Y: Revealing transcription factor and histone modification co-localization and dynamics across cell lines by integrating ChIP-seq and RNA-seq data. BMC Genomics. 19:9142018. View Article : Google Scholar

63 

Miguel V, Shaw IW and Kramann R: Metabolism at the crossroads of inflammation and fibrosis in chronic kidney disease. Nat Rev Nephrol. 21:39–56. 2025. View Article : Google Scholar

64 

Li Y, Min X, Zhang X, Cao X, Kong Q, Mao Q, Cheng H, Gou L, Li Y, Li C, et al: HSPA12A promotes c-Myc lactylation-mediated proliferation of tubular epithelial cells to facilitate renal functional recovery from kidney ischemia/reperfusion injury. Cell Mol Life Sci. 81:4042024. View Article : Google Scholar : PubMed/NCBI

65 

Xiang T, Wang X, Huang S, Zhou K, Fei S, Zhou B, Yue K, Li Q, Xue S, Dai Y, et al: Inhibition of PKM2 by shikonin impedes TGF-β1 expression by repressing histone lactylation to alleviate renal fibrosis. Phytomedicine. 136:1563242025. View Article : Google Scholar

66 

Zheng T, Gu YP, Wang JM, Huang TT, Gou LS and Liu YW: Lactate-triggered histone lactylation contributes to podocyte epithelial-mesenchymal transition in diabetic nephropathy in mice. Chem Biol Interact. 408:1114182025. View Article : Google Scholar : PubMed/NCBI

67 

Jia L, Sheng X, Zamperetti A, Xie Y, Corradi V, Chandel S, De Cal M, Montin DP, Caprara C and Ronco C: Combination of biomarker with clinical risk factors for prediction of severe acute kidney injury in critically ill patients. BMC Nephrol. 21:5402020. View Article : Google Scholar : PubMed/NCBI

68 

Legouis D, Ricksten S-E, Faivre A, Verissimo T, Gariani K, Verney C, Galichon P, Berchtold L, Feraille E, Fernandez M, et al: Altered proximal tubular cell glucose metabolism during acute kidney injury is associated with mortality. Nat Metab. 2:732–743. 2020. View Article : Google Scholar : PubMed/NCBI

69 

Sklienka P, Maca J, Neiser J, Bursa F, Sevcik P, Frelich M, Petejova N, Svagera Z, Tomaskova H and Zahorec R: Physiologic risk factors for early acute kidney injury in severely injured patients. Bratisl Lek Listy. 121:779–785. 2020.PubMed/NCBI

70 

Nasu T, Ueda K, Kawashima S, Okishio Y, Kunitatsu K, Iwasaki Y and Kato S: Prediction of early acute kidney injury after trauma using prehospital systolic blood pressure and lactate levels: A prospective validation study. Injury. 53:81–85. 2022. View Article : Google Scholar

71 

Gong C, Jiang Y, Tang Y, Liu F, Shi Y, Zhou H and Xie K: Elevated serum lactic acid level is an independent risk factor for the incidence and mortality of sepsis-associated acute kidney injury. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 34:714–720. 2022.In Chinese. PubMed/NCBI

72 

Flores-Salinas HE, Zambada-Gamboa AJ, Garcia-Garduño TC, Rodríguez-Zavala G, Valle Y, Chávez-Herrera JC, Martinez-Gutierrez PE, Godinez-Flores A, Jiménez-Limón S and Padilla-Gutiérrez JR: Association of postoperative serum lactate levels with acute kidney injury in mexican patients undergoing cardiac surgery. Clin Pract. 14:1100–1109. 2024. View Article : Google Scholar : PubMed/NCBI

73 

Wang Z, Xu J, Kang Y, Liu L, Zhang L and Wang D: Early dynamic behavior of lactate in predicting continuous renal replacement therapy after surgery for acute type A aortic dissection. Front Cardiovasc Med. 9:9486722022. View Article : Google Scholar : PubMed/NCBI

74 

Kahyaoglu M, Karaduman A, Geçmen Ç, Candan Ö, Güner A, Cakmak EO, Bayam E, Yılmaz Y, Çelik M, Izgi IA and Kirma C: Serum lactate level may predict the development of acute kidney injury in acute decompensated heart failure. Turk Kardiyol Dern Ars. 48:683–689. 2020.PubMed/NCBI

75 

Zhou X, He Y, Hu L, Zhu Q, Lin Q, Hong X, Huang W, Shan P and Liang D: Lactate level and lactate clearance for acute kidney injury prediction among patients admitted with ST-segment elevation myocardial infarction: A retrospective cohort study. Front Cardiovasc Med. 9:9302022022. View Article : Google Scholar : PubMed/NCBI

76 

Wang R, Wang S, Zhang J, He M and Xu J: Serum lactate level in early stage is associated with acute kidney injury in traumatic brain injury patients. Front Surg. 8:7611662021. View Article : Google Scholar

77 

Xu J, Ma X, Yu K, Wang R, Wang S, Liu R, Liu H, Gao H, Yu K and Wang C: Lactate up-regulates the expression of PD-L1 in kidney and causes immunosuppression in septic Acute Renal Injury. J Microbiol Immunol Infect. 54:404–410. 2021. View Article : Google Scholar

78 

Tan C, Gu J, Li T, Chen H, Liu K, Liu M, Zhang H and Xiao X: Inhibition of aerobic glycolysis alleviates sepsis-induced acute kidney injury by promoting lactate/Sirtuin 3/AMPK-regulated autophagy. Int J Mol Med. 47:192021. View Article : Google Scholar :

79 

Shen Y, Jiang L, Wen P, Ye Y, Zhang Y, Ding H, Luo J, Xu L, Zen K, Zhou Y and Yang J: Tubule-derived lactate is required for fibroblast activation in acute kidney injury. Am J Physiol Renal Physiol. 318:F689–F701. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Chu X, Di C, Chang P, Li L, Feng Z, Xiao S, Yan X, Xu X, Li H, Qi R, et al: Lactylated histone H3K18 as a potential biomarker for the diagnosis and predicting the severity of septic shock. Front Immunol. 12:7866662021. View Article : Google Scholar

81 

Guo W, Song Y, Sun Y, Du H, Cai Y, You Q, Fu H and Shao L: Systemic immune-inflammation index is associated with diabetic kidney disease in type 2 diabetes mellitus patients: Evidence from NHANES 2011-2018. Front Endocrinol (Lausanne). 13:10714652022. View Article : Google Scholar : PubMed/NCBI

82 

Roointan A, Gheisari Y, Hudkins KL and Gholaminejad A: Non-invasive metabolic biomarkers for early diagnosis of diabetic nephropathy: Meta-analysis of profiling metabolomics studies. Nutr Metab Cardiovasc Dis. 31:2253–2272. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Jiang C, Ma X, Chen J, Zeng Y, Guo M, Tan X, Wang Y, Wang P, Yan P, Lei Y, et al: Development of serum lactate Level-based nomograms for predicting diabetic kidney disease in type 2 diabetes mellitus patients. Diabetes Metab Syndr Obes. 17:1051–1068. 2024. View Article : Google Scholar : PubMed/NCBI

84 

Tang L, Yang Q, Ma R, Zhou P, Peng C, Xie C, Liang Q, Wu T, Gao W, Yu H, et al: Association between lactate dehydrogenase and the risk of diabetic kidney disease in patients with type 2 diabetes. Front Endocrinol (Lausanne). 15:13699682024. View Article : Google Scholar : PubMed/NCBI

85 

Muiru AN, Hsu JY, Zhang X, Appel LJ, Chen J, Cohen DL, Drawz PE, Freedman BI, Go AS, He J, et al: Risk for chronic kidney disease progression after acute kidney injury: Findings from the chronic renal insufficiency cohort study. Ann Intern Med. 176:961–968. 2023. View Article : Google Scholar : PubMed/NCBI

86 

Chesnaye NC, Ortiz A, Zoccali C, Stel VS and Jager KJ: The impact of population ageing on the burden of chronic kidney disease. Nat Rev Nephrol. 20:569–585. 2024. View Article : Google Scholar : PubMed/NCBI

87 

Tanemoto M: Gap acidosis except lactic acidosis develops and progresses during chronic kidney disease stage G5. Clin Exp Nephrol. 23:1045–1049. 2019. View Article : Google Scholar : PubMed/NCBI

88 

Zhao B, Xu Y, Chen Y, Cai Y, Gong Z, Li D, Kuang H, Liu X, Zhou H, Liu G and Yin Y: Activation of TRPV4 by lactate as a critical mediator of renal fibrosis in spontaneously hypertensive rats after moderate- and high-intensity exercise. Front Physiol. 13:9270782022. View Article : Google Scholar : PubMed/NCBI

89 

Nagy A, Pethő D, Gáll T, Zavaczki E, Nyitrai M, Posta J, Zarjou A, Agarwal A, Balla G and Balla J: Zinc Inhibits HIF-Prolyl Hydroxylase Inhibitor-Aggravated VSMC calcification induced by high phosphate. Front Physiol. 10:15842020. View Article : Google Scholar : PubMed/NCBI

90 

Huang Z, Liao Y, Zheng Y, Ye S, Zhang Q, Yu X, Liu X and Li N: Zinc deficiency causes glomerulosclerosis and renal interstitial fibrosis through oxidative stress and increased lactate metabolism in rats. Biol Trace Elem Res. 203:2084–2098. 2025. View Article : Google Scholar :

91 

Li M, Jia F, Zhou H, Di J and Yang M: Elevated aerobic glycolysis in renal tubular epithelial cells influences the proliferation and differentiation of podocytes and promotes renal interstitial fibrosis. Eur Rev Med Pharmacol Sci. 22:5082–5090. 2018.PubMed/NCBI

92 

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 Mede. 208:516–529. 2023. View Article : Google Scholar

93 

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

94 

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

95 

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

96 

Md Shakhih MF, Rosslan AS, Noor AM, Ramanathan S, Lazim AM and Wahab AA: Review-enzymatic and Non-enzymatic electrochemical sensor for lactate detection in human biofluids. J Electrochem Soc. 168:0675022021. View Article : Google Scholar

97 

Henry H, Marmy Conus N, Steenhout P, Béguin A and Boulat O: Sensitive determination of D-lactic acid and L-lactic acid in urine by high-performance liquid chromatography-tandem mass spectrometry. Biomed Chromatogr. 26:425–428. 2012. View Article : Google Scholar

98 

Bollella P, Sharma S, Cass AEG and Antiochia R: Microneedle-based biosensor for minimally-invasive lactate detection. Biosens Bioelectron. 123:152–159. 2019. View Article : Google Scholar

99 

Xie Y, Li K, Liu J, Zhou Y, Zhang C, Yu Y, Wang J, Su L and Zhang X: A smart lab on a wearable microneedle patch with convolutional neural network-enhanced colorimetry for early warning of syndrome of inappropriate antidiuretic hormone secretion. Aggregate. 6:e6712025. View Article : Google Scholar

100 

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

101 

Sun J, Dai W, Guo Q, Gao Y, Chen J, Chen JL, Mao G, Sun H and Peng YK: Self-powered wearable electrochemical sensor based on composite conductive hydrogel medium for detection of lactate in human sweat. Biosens Bioelectron. 277:1173032025. View Article : Google Scholar : PubMed/NCBI

102 

Chen Z, Zhu Z, Liang W, Luo Z, Hu J, Feng J, Zhang Z, Luo Q, Yang H and Ding G: Reduction of anaerobic glycolysis contributes to angiotensin II-induced podocyte injury with foot process effacement. Kidney Int. 103:735–748. 2023. View Article : Google Scholar : PubMed/NCBI

103 

Jiajun W, Kaifeng G and Jing Z: Urinary PKM2, a marker predicating acute kidney injury in patients with sepsis. Int Urol Nephrol. 56:3039–3045. 2024. View Article : Google Scholar : PubMed/NCBI

104 

Bertelsen LB, Hansen ESS, Sadowski T, Ruf S and Laustsen C: Hyperpolarized pyruvate to measure the influence of PKM2 activation on glucose metabolism in the healthy kidney. NMR Biomed. 34:e45832021. View Article : Google Scholar : PubMed/NCBI

105 

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

106 

Chen Y, Bai X, Chen J, Huang M, Hong Q, Ouyang Q, Sun X, Zhang Y, Liu J, Wang X, et al: Pyruvate kinase M2 regulates kidney fibrosis through pericyte glycolysis during the progression from acute kidney injury to chronic kidney disease. Cell Prolif. 57:e135482024. View Article : Google Scholar :

107 

Wang J, Ren Y, Zheng X, Kang J, Huang Z, Xu L and Wang Y: Anti-fibrotic effects of low toxic Microcystin-RR on Bleomycin-induced pulmonary fibrosis: A comparison with Microcystin-LR. Front Pharmacol. 12:6759072021. View Article : Google Scholar : PubMed/NCBI

108 

Ren Y, Wang J, Guo W, Chen J, Wu X, Gu S, Xu L, Wu Z and Wang Y: Renoprotection of Microcystin-RR in unilateral ureteral Obstruction-induced renal fibrosis: Targeting the PKM2-HIF-1α pathway. Front Pharmacol. 13:8303122022. View Article : Google Scholar

109 

Gong M, Guo Y, Dong H, Wu F, He Q, Gong J and Lu F: Modified Hu-lu-ba-wan protects diabetic glomerular podocytes via promoting PKM2-mediated mitochondrial dynamic homeostasis. Phytomedicine. 123:1552472024. View Article : Google Scholar

110 

Qian L, Ren S, Xu Z, Zheng Y, Wu L, Yang Y, Wang Y, Li J, Yan S and Fang Z: Qian yang yu yin granule improves renal injury of hypertension by regulating metabolic reprogramming mediated by HIF-1α/PKM2 positive feedback loop. Front Pharmacol. 12:6674332021. View Article : Google Scholar

111 

Wang M, Zeng F, Ning F, Wang Y, Zhou S, He J, Li C, Wang C, Sun X, Zhang D, et al: Ceria nanoparticles ameliorate renal fibrosis by modulating the balance between oxidative phosphorylation and aerobic glycolysis. J Nanobiotechnology. 20:32022. View Article : Google Scholar : PubMed/NCBI

112 

Fu X, Zhang J, Huang X, Mo Z, Sang Z, Duan W and Huang W: Curcumin antagonizes glucose fluctuation-induced renal injury by inhibiting aerobic glycolysis via the miR-489/LDHA pathway. Mediators Inflamm. 2021:61045292021. View Article : Google Scholar : PubMed/NCBI

113 

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

114 

Han W, Wang C, Yang Z, Mu L, Wu M, Chen N, Du C, Duan H and Shi Y: SRT1720 retards renal fibrosis via inhibition of HIF1A/GLUT1 in diabetic nephropathy. J Endocrinol. 241:85–98. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Venkatesan A, Roy A, Kulandaivel S, Natesan V and Kim SJ: p-Coumaric acid nanoparticles ameliorate diabetic nephropathy via regulating mRNA expression of KIM-1 and GLUT-2 in Streptozotocin-induced diabetic rats. Metabolites. 12:11662022. View Article : Google Scholar : PubMed/NCBI

116 

Chen X, Wang H, Jiang M, Zhao J, Fan C, Wang Y and Peng W: Huangqi (astragalus) decoction ameliorates diabetic nephropathy via IRS1-PI3K-GLUT signaling pathway. Am J Transl Res. 10:2491–2501. 2018.PubMed/NCBI

117 

Ritu, Xiong Y, Sharma HP, Goyal RK, Narwal S, Berwal A, Jain S, Priya M, Singh M, Agarwal G, et al: Bioflavonoid combination attenuates diabetes-induced nephropathy in rats via modulation of MMP-9/TIMP-1, TGF-β, and GLUT-4-associated pathways. Heliyon. 10:e332172024. View Article : Google Scholar

118 

Zhang J, Ding T, Zhang X, Tang D and Wang J: Dapagliflozin relieves renal injury in a diabetic nephropathy model by inducing autophagy through regulation of miR-30e-5p/AKT/mTOR pathway. Trop J Pharm Res. 21:2115–2123. 2022. View Article : Google Scholar

119 

Fatouros IG, Douroudos I, Panagoutsos S, Pasadakis P, Nikolaidis MG, Chatzinikolaou A, Sovatzidis A, Michailidis Y, Jamurtas AZ, Mandalidis D, et al: Effects of L-carnitine on oxidative stress responses in patients with renal disease. Med Sci Sports Exerc. 42:1809–1818. 2010. View Article : Google Scholar : PubMed/NCBI

120 

Sharma B and Yadav DK: L-Carnitine and chronic kidney disease: A comprehensive review on nutrition and health perspectives. J Pers Med. 13:2982023. View Article : Google Scholar : PubMed/NCBI

121 

Wen YK: Impact of acute kidney injury on metformin-associated lactic acidosis. Int Urol Nephrol. 41:967–972. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 56 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li X, Hu L, Hu Q and Jin H: Lactate and lactylation in the kidneys: Current advances and prospects (Review). Int J Mol Med 56: 121, 2025.
APA
Li, X., Hu, L., Hu, Q., & Jin, H. (2025). Lactate and lactylation in the kidneys: Current advances and prospects (Review). International Journal of Molecular Medicine, 56, 121. https://doi.org/10.3892/ijmm.2025.5562
MLA
Li, X., Hu, L., Hu, Q., Jin, H."Lactate and lactylation in the kidneys: Current advances and prospects (Review)". International Journal of Molecular Medicine 56.2 (2025): 121.
Chicago
Li, X., Hu, L., Hu, Q., Jin, H."Lactate and lactylation in the kidneys: Current advances and prospects (Review)". International Journal of Molecular Medicine 56, no. 2 (2025): 121. https://doi.org/10.3892/ijmm.2025.5562