Hostname: page-component-7857688df4-92hv7 Total loading time: 0 Render date: 2025-11-17T10:55:21.424Z Has data issue: false hasContentIssue false

The new insights of lactate in various kidney diseases

Published online by Cambridge University Press:  14 October 2025

Kexin Guan
Affiliation:
Department of Nephrology, Xijing Hospital, Air Force Medical University , Shaanxi, China
Yuzhan Zhang
Affiliation:
Department of Nephrology, Xijing Hospital, Air Force Medical University , Shaanxi, China
Shuxian Guo
Affiliation:
Department of Nephrology, Xijing Hospital, Air Force Medical University , Shaanxi, China
Xiaoxuan Ning
Affiliation:
Department of Geriatrics, Xijing Hospital, Air Force Medical University , Shaanxi, China
Shiren Sun*
Affiliation:
Department of Nephrology, Xijing Hospital, Air Force Medical University , Shaanxi, China
*
Corresponding author: Shiren Sun; Email: sunshiren@medmail.com.cn
Rights & Permissions [Opens in a new window]

Abstract

Background

Kidneys are crucial for systemic lactate homeostasis, and a proper lactate balance subsequently supports normal kidney structure and function. The physiological lactate production-clearance axis along the proximal-distal tubular network may represent an important mechanism for maintaining tubulointerstitial microenvironmental balance. In the context of kidney diseases, the dynamic changes in lactate levels reveal the process of renal metabolic remodelling and even participate in the regulation of disease occurrence and progression.

Methods

This review systematically combs the maintenance of renal lactate homeostasis under physiological conditions and integrates current research findings on the roles of lactate in the initiation and progression of various kidney diseases, as well as the underlying core molecular mechanisms.

Results

Existing studies confirm that, in a variety of kidney diseases, abnormal lactate levels are closely associated with the occurrence of renal metabolic remodelling, and lactate itself can further regulate the progression of kidney diseases. Targeted regulation of lactate metabolism or lactate-related mechanisms of action is expected to provide a new perspective for the treatment of kidney diseases.

Conclusion

The exploration of lactate-related mechanisms offers potential insights for developing novel strategies for early diagnosis and therapeutic intervention of kidney diseases; however, more in-depth studies are still required to translate these findings into clinical practice.

Information

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - NCCreative Common License - ND
This is an Open Access article, distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives licence (http://creativecommons.org/licenses/by-nc-nd/4.0), which permits non-commercial re-use, distribution, and reproduction in any medium, provided that no alterations are made and the original article is properly cited. The written permission of Cambridge University Press must be obtained prior to any commercial use and/or adaptation of the article.
Copyright
© The Author(s), 2025. Published by Cambridge University Press

Introduction

The kidney is a vital organ responsible for blood filtration, maintaining water and electrolyte balance, regulating acid-base equilibrium, controlling blood pressure and supporting endocrine functions, all of which are enabled by its unique structure, including the glomerular filtration barrier and tubular reabsorption/secretion systems. However, kidney diseases without effective therapeutic strategies pose a major global health threat, affecting more than 10% of the population, with conditions such as acute kidney injury (AKI), chronic kidney disease (CKD) and end-stage renal disease imposing significant socioeconomic burdens (Refs. 1, Reference Francis, Harhay, Ong, Tummalapalli, Ortiz, Fogo, Fliser, Roy-Chaudhury, Fontana, Nangaku, Wanner, Malik, Hradsky, Adu, Bavanandan, Cusumano, Sola, Ulasi and Jha2). Additionally, the incidence of renal cell carcinoma (RCC) has risen to 10–20 cases per 1,00,000 in Europe and America, with increasing rates in other regions (Ref. Reference Larcher3). Early detection and prevention remain key challenges in nephrology.

Risk factors for kidney diseases include both genetic and non-genetic influences. Genetic factors play a key role, encompassing single-gene mutations causing hereditary kidney diseases and polygenic risks arising from multiple gene loci. Advances in genomics have highlighted the association between genetic polymorphisms – particularly single-nucleotide polymorphisms – and kidney disease susceptibility, offering insights for early prediction, risk stratification and personalised treatment (Ref. Reference Jefferis, Hudson, Lacaze, Bakshi, Hawley, Patel and Mallett4). Nongenetic contributors include infections, nephrotoxic drug/environmental toxin exposure and metabolic disorders, which significantly impact disease onset and progression. Although research has advanced, many underlying mechanisms remain unclear.

For a long time after its discovery in 1780, lactate was regarded merely as a glycolytic ‘metabolic waste’ under hypoxic conditions, with no major physiological role and was even harmful in excess (Ref. Reference Haas, Cucchi, Smith, Pucino, Macdougall and Mauro5). However, the 1980s lactate shuttle hypothesis revealed its crucial function, demonstrating that lactate shuttles between cells and contributes significantly to the physiological microenvironment (Refs. Reference Brooks6, Reference Brooks7). As both a metabolic substrate and signalling molecule, lactate directly participates in glucose metabolism, intercellular communication and other processes and plays a key role in maintaining organismal homeostasis (Refs. Reference Zhang, Peng, Zheng, Yang, Zhang, Ma, Qin, Qin and Zheng8, Reference Certo, Tsai, Pucino, Ho and Mauro9). The kidneys play a crucial role in maintaining the balance of lactate in the body; however, in the past, it seemed that the relationship between lactate and the kidney was limited to a simple association between consumption and excretion. With advances in the understanding of lactate levels, its role in maintaining renal homeostasis and contributing to pathological conditions has become increasingly clear. Moreover, given that the kidneys are highly metabolically active organs that primarily rely on fatty acid oxidation for energy under normal physiological conditions, maintaining homeostasis in energy metabolism is essential for their proper function. Recent studies have shown that in response to injuries, such as ischaemia, hypoxia and toxins, kidney energy metabolism shifts towards glycolysis (Refs. Reference Chrysopoulou and Rinschen10, Reference van der Rijt, Leemans, Florquin, Houtkooper and Tammaro11). This shift leads to local lactate accumulation, significantly contributing to the onset and progression of various diseases (Refs. Reference Miguel, Shaw and Kramann12Reference Zhu, Hu, Chen, Feng, Yang, Liang and Ding14). Notably, in many relevant clinical studies, the levels of lactate and several key molecules involved in metabolic pathways are often described as prognostic indicators in kidney diseases, with suggestive significance. Additionally, a number of gene-level studies and analyses have also indicated that lactate metabolism-related genes may play a role in the occurrence and progression of kidney diseases. Despite this, the specific regulatory network of lactate metabolism – its role in various renal physiological processes, alterations under pathological conditions and contributions to the onset and progression of renal diseases – remains incompletely understood the precise mechanisms linking lactate metabolism to renal disease initiation and progression remain unclear. This review systematically elucidates the maintenance of lactate homeostasis within the kidney and describes the role of lactate in the pathogenesis and progression of kidney diseases. Furthermore, this study provides an in-depth synthesis of the underlying molecular mechanisms, holding promise for offering novel perspectives on therapeutic interventions for kidney diseases.

Lactate metabolism and its homeostasis in the kidney

In glucose oxidative metabolism, glucose is gradually catabolised to pyruvate when it enters the cytoplasm for oxidation and utilisation, and pyruvate can be further converted into acetyl-CoA to enter the tricarboxylic acid (TCA) cycle or directly reduced to lactate under the action of lactate dehydrogenase (LDH) according to the sufficiency of oxygen (Ref. Reference Shen, Jiang, Wen, Ye, Zhang, Ding, Luo, Xu, Zen, Zhou and Yang15). When the demand for oxygen and ATP exceeds the cellular supply, the TCA cycle is inhibited and the glycolytic pathway is activated to replenish the supply of ATP, resulting in increased lactate production (Refs. Reference Rabinowitz and Enerback16, Reference Li, Yang, Zhang, Lin, Fu, An, Zou, Wang, Wang and Yu17). Although it is a nontoxic molecule and can be oxidised to release energy, the excessive accumulation of lactate could lead to a dangerous increase in the concentration of hydrogen ions (H+) concentration, indicating a change in acid–base homeostasis (Ref. Reference De Corte18). Rapid clearance of lactate from tissues and the circulation to maintain balance is therefore important. As one of the primary organs for systemic lactate clearance, the kidney mediates this process mainly via cellular uptake and metabolic processing, not through urinary excretion (Ref. Reference Bellomo19). The clearance of lactate in cells is mainly through the following two catabolic pathways. In the first pathway, lactate is oxidised to pyruvate under the catalysis of LDH. Then, pyruvate enters mitochondria and is metabolised by the TCA cycle, and irreversible lactate removal is achieved by pyruvate dehydrogenase (PDH) (Ref. Reference Brooks20). Another pathway involves the conversion to glucose through gluconeogenesis (Refs. Reference Rabinowitz and Enerback16, Reference Li, Yang, Zhang, Lin, Fu, An, Zou, Wang, Wang and Yu17). It can be concluded that a close relationship exists between the level of lactate in the body and LDH and PDH expression and activity. Additionally, different monocarboxylate transporters (MCTs) provide the conditions for the transport of lactate between various cells, and their distribution depends on the cell type (Ref. Reference Wu, Zhang, Khan, Wu, Pandupuspitasari, Tang, Guan, Sun and Huang21). A comprehensive summary of will be conducted on the pivotal molecules within the renal system that play crucial roles in lactate production and clearance, as well as its transportation, is provided below (Figure 1).

Figure 1. Lactate transport and metabolism in the kidney under physiological and pathological circumstances. LDH and PDH coordinate the production and clearance of lactate, while MCTs facilitate lactate transport into or out of cells, collectively maintaining lactate homeostasis in the kidney. MCTs: monocarboxylate transporters; LDH: lactate dehydrogenase; PDH: pyruvate dehydrogenase; TCA: tricarboxylic acid. (Figure was created with Biorender.com.)

LDH and PDH – maintaining homeostasis of lactate production and metabolism

LDH is among key rate-limiting enzymes that catalyse the bidirectional conversion of pyruvate and lactate in glycolysis. It is a tetramer composed of different subunits LDHA and LDHB (Ref. Reference Frank, Raue, Fuhrmann, Sirait-Fischer, Reuse, Weigert, Lütjohann, Hiller, Syed and Brüne22). LDHA is mainly responsible for converting pyruvate into lactate and NAD. In contrast, LDHB converts lactate into pyruvate to promote oxidative metabolism and the proportion of the two in the tetramer determines the overall direction of the reaction (Refs. Reference Certo, Tsai, Pucino, Ho and Mauro9, Reference Yang, Luo, Zhao, Li, Wang, Zeng, Yang, Zheng, Jie, Kang, Li, Liu, Zhou and Liu23). Previous studies have shown that LDHA is mainly expressed mainly in the proximal nephron segment, whereas LDHB is mainly expressed mainly in the distal nephron under normal physiological conditions, suggesting that the proximal tubule is more inclined to produce lactate, whereas the distal tubule is more inclined to utilize lactate physiologically (Ref. Reference Osis, Traylor, Black, Spangler, George, Zarjou, Verlander and Agarwal24). Similarly, single-cell RNA-sequencing (scRNA-seq) datasets derived from Wu and Malone et al. as well as Wu and Uchimura et al. revealed that Ldha mRNA expression was greater in the proximal tubules (PTs) than in the distal tubules, whereas Ldhb mRNA expression was higher in the distal nephrons of the loop of Henle and distal convoluted tubules than in the PTs (Refs. Reference Wu, Malone, Donnelly, Kirita, Uchimura, Ramakrishnan, Gaut and Humphreys25, Reference Wu, Uchimura, Donnelly, Kirita, Morris and Humphreys26). Moreover, exposure to injurious stimuli induces corresponding alterations in LDH expression. Analysis of the single-nucleus RNA-sequencing (scRNA-seq) datasets in this study revealed upregulation of the expression of both Ldha and Ldhb gene transcripts following ischaemia–reperfusion injury (IRI), highlighting the initial metabolic shift towards glycolysis postinjury (Ref. Reference Osis, Traylor, Black, Spangler, George, Zarjou, Verlander and Agarwal24). Interestingly, LDHA and LDHB expression in different nephron segments displays spatial and time-dependent responses to ischaemic injury. Both LDHA and LDHB decrease after ischaemic acute kidney injury and recover later; LDHA decreases early (Day 1), followed by a decrease in LDHB on Day 3 postinjury. in vitro experiments conducted by Osis et al. revealed a significant upregulation of LDHA expression in metanephric tubular epithelial cells (TECs) under hypoxic conditions, whereas LDHB levels remained unchanged. Notably, distal nephron expression of LDHB increased between 3 and 7 days after injury but normalised by 14 days. These data suggest that proximal tubules are lactate producers, whereas distal nephron segments are lactate consumers regardless of whether they are in a physiological state or when confronted with stress dependency on LDH subunits and differential expression among distinct renal units. However, several studies have reported a reduction in LDH protein expression in the kidney following the establishment of mouse models simulating unilateral or bilateral renal ischaemia-reperfusion, whereas concurrent elevations in plasma and urine LDH levels have been noted (Refs. Reference Osis, Traylor, Black, Spangler, George, Zarjou, Verlander and Agarwal24, Reference Zager, Johnson and Becker27, Reference Zager, Johnson and Becker28). This phenomenon could be attributed to the release of LDH from damaged cells into the bloodstream or urine (Ref. Reference Zager, Johnson and Becker28).

PDH facilitates the irreversible decarboxylation of pyruvate to acetyl-CoA, thereby regulating carbon entry into the TCA cycle and indirectly facilitating lactate clearance (Refs. Reference Jha and Suk29, Reference Jha, Lee and Suk30). It functions as a pivotal dehydrogenase enzyme that regulates mitochondrial metabolic pathways, connects glycolysis and the TCA cycle and plays a critical role in cellular metabolism. In the case of hypoxia or mitochondrial damage, PDH inhibition leads to reduced lactate clearance, accompanied by a compensatory increase in lactate production, thereby resulting in abnormally elevated lactate levels in the microenvironment and circulation (Refs. Reference Luengo, Li, Gui, Sullivan, Zagorulya, do, Ferreira, Naamati, Ali, Lewis, Thomas, Spranger, Matheson and Vander Heiden31, Reference Kamel, Oh and Halperin32). When cisplatin-induced kidney injury occurs, there is an increase in PDH phosphorylation increases, leading to PDH deactivation (Ref. Reference Galgamuwa, Hardy, Dahlstrom, Blackburn, Wium, Rooke, Cappello, Tummala, Patel, Chuah, Tian, McMorrow, Board and Theodoratos33). Similarly, Lan et al. reported that PDH phosphorylation was upregulated in the kidneys of IRI mice, concomitant with the accumulation of lactate (Ref. Reference Lan, Geng, Singha, Saikumar, Bottinger, Weinberg and Venkatachalam34). The reduced levels of PDH during kidney injury may impede the conversion of pyruvate to acetyl-CoA, thereby compromising the clearance of lactate (Ref. Reference Zager, Johnson and Becker27).

Monocarboxylate transporters – maintaining equilibrium of lactate transport

The transport of lactate across the plasma membrane occurs through two selective transporters, namely proton-coupled monocarboxylate transporters (MCT1–4) and sodium-coupled monocarboxylate transporters (SMCT1–2) (Refs. Reference Yanase, Takebe, Nio-Kobayashi, Takahashi-Iwanaga and Iwanaga35Reference Certo, Llibre, Lee and Mauro37). The homeostatic balance of lactate in different tissue microenvironments can be maintained through the expression and activity of these monocarboxylate transporters (Refs. Reference Rabinowitz and Enerback16, Reference Becker, Mohebbi, Perna, Ganapathy, Capasso and Wagner38). MCTs need to bind to glycosylated helper proteins to ensure correct expression on the plasma membrane for normal transport protein activity (Ref. Reference Ippolito, Morandi, Giannoni and Chiarugi39). In the transport process, free protons first bind to MCT, and then bind to lactate and then conformational changes occur in the transporter to a ‘closed’, state, where protons and lactate are exposed on the other side of the membrane and the release of protons is followed by the release of lactate. When MCT loses protons, it undergoes a conformational change that restores its initial structure before the next transport (Refs. Reference Li, Yang, Zhang, Lin, Fu, An, Zou, Wang, Wang and Yu17, Reference Halestrap36). In general, MCTs with high substrate affinity, such as MCT2 (Km ≈ 0.7 mM), is predominantly expressed in lactate-consuming tissues (e.g., the brain and heart), where they facilitate efficient cellular lactate uptake. In contrast, MCTs with lower affinity, such as MCT4 (Km ≈ 35 mM), are primarily found in highly glycolytic cells (e.g., astrocytes), enabling rapid lactate export to maintain glycolytic flux (Ref. Reference Sheikh-Hamad40). Owing to their different affinities for lactate, MCTs play disparate roles in both the uptake and efflux of lactate from cells, contingent upon the concentration gradient of this metabolite (Ref. Reference Certo, Llibre, Lee and Mauro37). However, research on SMCTs has long been relatively scarce. Lactate transport mediated by SMCT1–2 primarily facilitates the reabsorption of lactate by tissues, so as to maintain the normal physiological functions of the body (Ref. Reference Iwanaga and Kishimoto41). Thus, MCTs and SMCTs are jointly responsible for lactate homeostasis and the impairment or inactivation of transporters leads to compromised lactate transportation and functionality.

In the kidney, MCT1 (Km ≈ 3.5–10 mM) has been detected in proximal renal tubular cells, whereas MCT2 is predominantly localised in the thick ascending limb of the loop of Henle and the distal convoluted tubule under unperturbed physiological conditions, as reported (Refs. Reference Becker, Mohebbi, Perna, Ganapathy, Capasso and Wagner38, Reference Sheikh-Hamad40). MCT4, a potent lactate output channel, that is expressed in glycolytic cells and induced by hypoxia and redox signalling, has been poorly studied in normal kidneys (Ref. Reference Ippolito, Morandi, Giannoni and Chiarugi39). The tissue distribution of MCT3 is limited, with no detectable expression observed in the kidney. Using the renal transcriptomics database (Nephroseq), we analysed the expression levels of SLC16A1 (encoding MCT1), SLC16A7 (encoding MCT2) and SLC16A3 (encoding MCT4) in a microarray dataset consisting of kidney biopsy specimens from patients with CKD, as constructed by Shunsaku Nakagawa et al. (Ref. Reference Nakagawa, Nishihara, Miyata, Shinke, Tomita, Kajiwara, Matsubara, Iehara, Igarashi, Yamada, Fukatsu, Yanagita, Matsubara and Masuda42). The results showed greater SLC16A1, SLC16A7 and SLC16A3 expression in the kidneys of CKD patients (n = 53) than in those of normal patients (n = 8; Figure 2). Despite not being a specific lactate transport receptor, these findings still have implications for the enhancement of lactate-mediated communication in the kidneys of patients with CKD. In addition, both SMCT1 and SMCT2 are found to be expressed in the proximal tubule (Ref. Reference Gopal, Umapathy, Martin, Ananth, Gnana-Prakasam, Becker, Wagner, Ganapathy and Prasad43). Researchers utilize c/ebpδ−/− mice, a model for the kidney-specific double knockout of SLC5A8 (encoding SMCT1) and SLC5A12 (encoding SMCT2) and observe a significant upregulation of urine lactate, whereas blood lactate is downregulated compared with that in wild-type mice (Ref. Reference Thangaraju, Ananth, Martin, Roon, Smith, Sterneck, Prasad and Ganapathy44). These findings suggest that the majority of lactate reabsorption in the kidney is attributed to SMCTs. MCT-mediated lactate transport establishes a communication pathway between cells and provides a proper condition for lactate homeostasis and the balance between glycolysis and gluconeogenesis in the kidney.

Figure 2. The expression of MCT1, MCT2 and MCT4 exhibit significant upregulation in the kidneys of patients with CKD. The mRNA levels of SLC16A1, SLC16A7 and SLC16A3 in kidney specimens of CKD (n = 53 samples) and control (n = 8 samples) in GSE66494 dataset are showed. Unpaired t-test, p < 0.05.

In summary, the kidneys serve as both producers and consumers of lactate. Considering the anatomical proximity of these segments, there is a conceivably efficient ‘lactate production-clearance axis’ through the spatiotemporal-specific expression of LDH/PDH and MCTs in proximal tubules and distal tubules to preserve intrarenal lactate equilibrium (Figure 3), similar to the lactate shuttle existing in the brain or the transfer from skeletal muscle to the heart during physical exercise (Ref. Reference Brooks20). Specifically, lactate is primarily generated in the proximal nephron and is subsequently transported to the distal nephron, where it is taken up by cells to meet their energy requirements or for signalling. However, on the basis of the conclusions of other previous studies that the lactate concentration in the medulla is significantly greater than that in the cortex in terms of the distribution of lactate in the kidney and that the gluconeogenesis of lactate in the kidney is performed mainly by the proximal tubule, another hypothesis has been proposed (Refs. Reference Legouis, Faivre, Cippà and de Seigneux45, Reference Bankir46). According to this hypothesis, the medulla mainly generates lactate through glycolysis, whereas the cortex is responsible for absorbing and oxidising lactate to generate energy or synthesising glucose through the gluconeogenesis pathway and then rereleasing this glucose back to the medulla to participate in glycolysis and energy metabolism. To more thoroughly clarify the metabolic fate of lactate in the kidneys, an important physiological process, it is necessary to conduct more systematic and extensive research. These findings provide a new theoretical basis for the study of renal physiology and may offer scientific evidence for the diagnosis and treatment strategies of related diseases.

Figure 3. Schematic Diagram of the "lactate production-clearance axis" between the proximal and distal renal tubules. Under physiological conditions of the kidney, lactate is primarily generated in the proximal nephron under the catalysis of LDHA and is subsequently transported to the distal nephron via MCTs, where it is taken up by cells to meet their energy requirements or for signalling. (Figure was created with Biorender.com.)

Disease

Lactate homeostasis plays a critical role in renal function and systemic internal balance. Therefore, exploring the alterations in lactate homeostasis in the kidney under pathological conditions and its potential contribution to the pathogenesis and progression of kidney diseases is imperative. Recent studies have yielded significant insights into this area, with several key findings summarised below.

Genetic susceptibility

Genome-wide association studies (GWAS) can be used to identify potential associations between genetic regions in the genome and diseases. Large-scale GWAS datasets have shown that loci of lactate metabolism-related genes, namely GCKR (rs1260326) and PFKFB2 (rs2808454), are associated with eGFRcrea, suggesting a possible link to the risk of renal function impairment (Refs. Reference Wuttke47Reference Liu, Doke, Guo, Sheng, Ma, Park, Vy, Nadkarni, Abedini, Miao, Palmer, Voight, Li, Brown, Ritchie, Shu and Susztak49). In addition, existing studies indicate that specific genetic backgrounds may enhance the susceptibility to lactate metabolism disorders in individuals with kidney diseases. When renal pathological damage is present, genetic factors synergistically exacerbate the risk of local or systemic lactate accumulation. Guo et al. developed a prognostic model for clear cell renal cell carcinoma based on lactate metabolism- and transport-related genes (PNKD, SLC16A8 and SLC5A8), demonstrating that genetic susceptibility contributes to disease progression through lactate metabolic dysregulation, remodelling of the immunosuppressive tumour microenvironment and activation of cell cycle signalling pathways (Ref. Reference Guo, Zhang, Wang, Yuan, Tang, Zhang, Chen and Wang50). Subsequent studies further constructed analogous prognostic models for renal cancer by identifying lactate-associated gene signatures, revealing that genetic heterogeneity mediates immunosuppressive microenvironment formation and therapeutic resistance via lactate metabolism reprogramming (Refs. Reference Sun, Tao, Guo, Jing, Zhang, Wang, Kong, Suo, Jiang and Wang51, Reference Wu, Wu, Sun, You, Zhang and Zhao52). However, current investigations into susceptibility to lactate metabolism disorders in kidney diseases predominantly focus on a limited number of candidate genes, with a notable absence of systematic research approaches, making it difficult to fully unravel the underlying genetic association mechanisms. The development of a genetic risk model for renal lactate metabolism disorder holds significant potential in improving risk stratification and enabling personalised management strategies for high-risk patients.

Hyperlactatemia and kidney

Hyperlactatemia refers to the accumulation of lactate in the blood due to excessive production or insufficient clearance of lactate and lactateosis refers to increased lactate accompanied by metabolic acidosis (Refs. Reference Rabinowitz and Enerback16, Reference Bellomo19). Lactateosis is the most common cause of metabolic acidosis in severe patients and elevated blood lactate concentrations have important effects on patient prognosis and even mortality (Refs. Reference Kamel, Oh and Halperin32, Reference Sun, Li, Chen and Qian53). Acidosis increases lactate metabolism in the kidneys. In the condition of hyperlactatemia, the normal function of the kidneys contributes to lactate clearance, mostly through lactate metabolism rather than excretion (Ref. Reference Madias54). This also means that in the case of lactate overproduction in the body, the kidney will bear a greater burden to maintain acid–base balance and the kidney will be more prone to lactateosis when it is dysfunctional. Lactateosis is often associated with AKI. When AKI occurs in patients, the consumption and clearance of lactate from the kidneys is reduced and impaired, which may lead to the accumulation of lactate in the blood (Refs. Reference Wen, Li, Li, Hu, Wei and Dong55, Reference Legouis, Ricksten, Faivre, Verissimo, Gariani, Verney, Galichon, Berchtold, Feraille, Fernandez, Placier, Koppitch, Hertig, Martin, Naesens, Pugin, McMahon, Cippà and de Seigneux56). From another standpoint, when the production of lactate increases because of lesions in other tissues in the body, the associated inflammatory mechanisms may lead to reduced lactate clearance by the kidney as well (Ref. Reference Kamel, Oh and Halperin32). Numerous clinical studies have consistently demonstrated a positive association between serum lactate levels and the occurrence of AKI during surgical procedures and severe trauma (Refs. Reference García, Manzano-Nunez, Bayona, Naranjo, Villa, Moreno, Ossa, Martinez, Martinez and Puyana57Reference Choi60). In other words, the blood lactate concentration can serve as an independent risk factor for AKI, and plays a pivotal warning role in early disease detection. More importantly, using a two-sample Mendelian randomisation analysis, we recently reported a causal relationship between hyperlactatemia and a broad range of CKD outcomes indicating that serum lactate levels may also be risk factors for CKD.

Kidney injury and fibrosis

When kidney injury occurs, there is a shift in energy metabolism within the kidney, leading to an increase in the activity of the glycolytic pathway, which contributes to the provision of energy and subsequently mitigates TEC injury (Refs. Reference Zhu, Hu, Chen, Feng, Yang, Liang and Ding14, Reference Lan, Geng, Singha, Saikumar, Bottinger, Weinberg and Venkatachalam34). Enhancement of glycolysis has been demonstrated in various models of AKI, including those induced by ischaemia-reperfusion, sepsis, folic acid, cisplatin and so on (Refs. Reference Shen, Jiang, Wen, Ye, Zhang, Ding, Luo, Xu, Zen, Zhou and Yang15, Reference Lan, Geng, Singha, Saikumar, Bottinger, Weinberg and Venkatachalam34, Reference Gómez, Kellum and Ronco61, Reference Xie, He, Zhang, Xu, Wen, Cao, Zhou, Luo, Yang and Jiang62). This is accompanied with by the accumulation of lactate, the end product of glycolysis, accumulated within damaged renal tubular cells and the tubulointerstitial microenvironment (Ref. Reference Shen, Jiang, Wen, Ye, Zhang, Ding, Luo, Xu, Zen, Zhou and Yang15). Considering the short-term replenishment of energy supply by lactate during injury, it can be postulated that lactate may have a protective effect on kidney injury at this stage, as evidenced by our results that lactate improves tubule injury and subsequent fibrosis after preoperative peritoneal injection of sodium lactate (Figure 4). Interestingly, Zeng et al. reported that both PDHA1 hyperacetylation-mediated lactate overproduction and intraperitoneal injection of exogenous sodium lactate before caecal ligation and puncture procedure promote sepsis-induced acute kidney injury (SAKI), indicating lactate plays a negative role in the AKI process (Ref. Reference An, Yao, Hu, Wu, Li, Li, Wu, Sun, Deng, Zhang, Gong, Huang, Chen and Zeng63). These studies suggest that the effects of lactate may differ in various types of AKI, and more research is necessary to elucidate the role of lactate in acute kidney injury.

Figure 4. The histopathological alterations in renal injury and fibrosis following lactate injection in the UIRI mouse model. LA: lactate intraperitoneal injection. Scale bar, 100 μm.

Although glycolysis initially enhances survival in TECs, sustained activation of glycolysis may contribute to progressive renal injury. Previous findings indicate that lactate generated through tubular glycolysis during the reparative phase of AKI has possess potential cytotoxic effects, thereby directly contributing to fibrogenesis (Refs. Reference Shen, Jiang, Wen, Ye, Zhang, Ding, Luo, Xu, Zen, Zhou and Yang15, Reference Faivre, Verissimo, Auwerx, Legouis and de Seigneux64Reference Yang, Huo, Cai, Zhang, Dong, Asara and Wei66). We examined the histopathological features of the kidneys of UIRI mice treated with continuous sodium intraperitoneal injection of sodium lactate and discovered that compared with those in the UIRI-saline group, the mice in the UIRI-lactate group had more obvious tubulointerstitial injury and more severe fibrosis (Figure 4). Notably, Shen et al. Injected the glycolysis inhibitors, 2-DG and oxamate, into mice with folate-induced AKI and reported that the lactate level in the renal tubules of these mice decreased significantly, whereas the fibrous deposition decreased. However, more morphological damage occurred in renal tubules (Ref. Reference Shen, Jiang, Wen, Ye, Zhang, Ding, Luo, Xu, Zen, Zhou and Yang15). These findings suggest that inhibiting the production of lactate by tubules could provide a certain degree of protection against fibrosis but at the same time may aggravate the mean renal injury in AKI (Refs. Reference Shen, Jiang, Wen, Ye, Zhang, Ding, Luo, Xu, Zen, Zhou and Yang15, Reference Yang, Huo, Cai, Zhang, Dong, Asara and Wei66, Reference Tan, Gu, Li, Chen, Liu, Liu, Zhang and Xiao67). Consistent with our previous study, we treated UIRI mice with 2-DG and found that 2-DG protected them from renal tubule interstitial fibrosis. It can be inferred that the dysregulation of lactate production and clearance in the kidney is intricately associated with the accelerated progression of renal fibrosis during the transition from the acute to chronic phase. The moderate inhibition of aerobic glycolysis is an effective approach for suppressing renal fibrosis, offering a promising therapeutic strategy for the treatment of AKI and CKD (Refs. Reference Ding, Jiang, Xu, Bai, Zhou, Yuan, Luo, Zen and Yang68, Reference Verissimo, Faivre, Rinaldi, Lindenmeyer, Delitsikou, Veyrat-Durebex, Heckenmeyer, Fernandez, Berchtold, Dalga, Cohen, Naesens, Ricksten, Martin, Pugin, Merlier, Haupt, Rutkowski, Moll, Cippà, Legouis and de Seigneux69). At present, the role of lactate in renal fibrosis is not completely clear, and it is urgent to elucidate the exact new mechanism of its activity to identify new therapeutic targets.

Diabetic kidney disease

As one of the leading causes of renal failure worldwide, diabetic kidney disease (DKD) is generally considered to be associated with altered cellular metabolism (Ref. Reference Sas, Kayampilly, Byun, Nair, Hinder, Hur, Zhang, Lin, Qi, Michailidis, Groop, Nelson, Darshi, Sharma, Schelling, Sedor, Pop-Busui, Weinberg, Soleimanpour, Abcouwer, Gardner, Burant, Feldman, Kretzler, Brosius and Pennathur70). Pathologically induced aberrant glycolysis also occurs in DKD and may play a key role in kidney injury (Refs. Reference Wu, Dong, Atefi, Liu, Elshimali and Vadgama71Reference Srivastava, Li, Kitada, Fujita, Yamada, Goodwin, Kanasaki and Koya74). Specifically, increased upregulation of IGFBP5 expression induces glycolysis, leading to increased renal inflammation and fibrosis, ultimately contributing to the progression of DKD progression (Ref. Reference Song, Wang, Fu, Chi, Geng, Liu, Cai, Chen, Wu and Hong75). Additionally, the formation and activity of the pyruvate kinase (PK) M2 (PKM2) tetramer are decreased in diabetic nephropathy, whereas the assembly of PKM2 plays a pivotal role in facilitating the accumulation of hypoxia-inducible factor 1α, mediating aberrant glycolysis and inducing fibrosis in DKD (Ref. Reference Qi, Keenan, Li, Ishikado, Kannt, Sadowski, Yorek, Wu, Lockhart, Coppey, Pfenninger, Liew, Qiang, Burkart, Hastings, Pober, Cahill, Niewczas, Israelsen, Tinsley, Stillman, Amenta, Feener, Vander Heiden, Stanton and King76). The findings from these studies suggest that the presence of aberrant glycolysis is linked to an unfavourable prognosis in patients with DKD. We previously reported that the production of lactate, the main metabolite of this process, is closely related to the expression of lactate dehydrogenase and Azushima et al. reported that the expression of Ldha and Ldhb isoforms is significantly increased in the kidneys of diabetic nephropathy (DN) mice. In particular, Ldha, the main enzyme that catalyses the conversion of pyruvate to lactate, changed most significantly in the proximal renal tubules (Refs. Reference Azushima72, Reference Schlosser77). Consistent with these findings, all the relevant clinical analyses have shown that the blood lactate level and urinary lactate levels significantly increase in DKD patients (Refs. Reference Wu, Dong, Atefi, Liu, Elshimali and Vadgama71, Reference Blantz78). Moreover, in recent studies, the significant increase in urinary lactate levels in diabetic nephropathy patients has been consistently associated with albuminuria, a remarkable feature of early kidney injury, suggesting that lactate metabolism may be a new biomarker for DKD (Refs. Reference Sas, Kayampilly, Byun, Nair, Hinder, Hur, Zhang, Lin, Qi, Michailidis, Groop, Nelson, Darshi, Sharma, Schelling, Sedor, Pop-Busui, Weinberg, Soleimanpour, Abcouwer, Gardner, Burant, Feldman, Kretzler, Brosius and Pennathur70, Reference Azushima72, Reference Chou, Lee, Chen, Hsieh, Huang, Li and Lee79). Lactate is known to accompany DKD, but whether lactate is also an indicator of DKD has not yet been determined. In addition to revealing the correlation between high lactate levels in the kidney and kidney damage, this undoubtedly provides a new therapeutic idea for diabetic nephropathy, that is, lactate metabolism may be a new intervention point.

Renal cell carcinoma

Proliferating tumour cells usually exhibit enhanced metabolic activity and obtain more energy rapidly through aerobic glycolysis (Warburg effect). In this case, the production of a large amount of lactate not only leads to the acidification of the tumour microenvironment, but also plays a role in tumour migration, invasion and immunosuppression (Ref. Reference Ippolito, Morandi, Giannoni and Chiarugi39). RCC is a typical malignant tumour that exhibits the Warburg effect (Ref. Reference Miranda-Gonçalves, Lameirinhas, Henrique, Baltazar and Jerónimo80). Moreover, lactate produced by glycolysis may increase the aggressiveness of RCC by reducing SIRT1 activity, and affecting and regulating the phenotype of tumour cell and adjacent normal cells, and thus accelerating tumour progression (Ref. Reference Miranda-Goncalves81). Beyond this, research has indicated that kidney cancer cells can promote tumour progression by breaking down fat around the kidney to produce lactate (Ref. Reference Wei82). Recent studies have focused on the role of lactate-related metabolism in the occurrence and development of renal cell carcinoma. The upregulated expression of LDHA is once again regarded as a potential indicator of poor prognosis in RCC patients (Refs. Reference Li, du, Li and Peng83, Reference Girgis, Masui, White, Scorilas, Rotondo, Seivwright, Gabril, Filter, Girgis, Bjarnason, Jewett, Evans, al-Haddad, Siu and Yousef84). Understanding the role of lactate itself in RCC and the underlying mechanism is necessary.

Potential mechanism of lactate in kidney disease

The occurrence of these common kidney disorders is typically concomitant with heightened glycolytic activity, resulting in an increase in the lactate concentration (Figure 5A). Given that lactate can be regarded as a potential risk factor or prognostic indicator for the onset of disease, we must not overlook its potential role and mechanism in the progression of kidney disease to effectively manage this condition (Figure 5B).

Figure 5. Lactate metabolism disorders in prevalent renal diseases and potential mechanisms of lactate’s involvement in those diseases’ progression. A. Elevated lactate concentration has been observed in the development of numerous renal diseases, yet its fluctuations and significance in the progression from AKI to CKD warrant further investigation. B. The occurrence of the above-mentioned diseases leads to alterations in the renal microenvironment due to increased lactate secretion. The diagram provides an overview of the potential mechanism by which lactate contributes to the progression of these diseases. Lactate is capable of directly facilitating the interaction between kidney cells, by activating specific signaling pathways, or entering cells for post-translational modifications to impact gene expression. GPR81: G-protein-coupled receptor 81; TRPV4: transient receptor potential vanilloid 4. MCT: monocarboxylate transporter; SIRT: sirtuin; HDAC: Histone deacetylase. (Figure was created with BioRender.com.)

Crosstalk between cells

Renal cells are capable of intercellular communication as well as communication with other cell types resident in the renal tissue (Ref. Reference Ebefors and Nyström85). The crosstalk plays a pivotal role in the exchange of signals in regulation renal physiological and pathological processes and is generally mediated by an array of bioactive substances, such as cytokine, exosome and metabolites, of which lactate is one. Studies using a folic acid mouse model of AKI have demonstrated that augmented tubular lactate excretion contributes to the activation of renal fibroblasts, which have been considered to have a pro-regenerative role in AKI (Refs. Reference Shen, Jiang, Wen, Ye, Zhang, Ding, Luo, Xu, Zen, Zhou and Yang15, Reference Comstock and Udenfriend65, Reference Guo, Cui, Jiao, Yao, Zhao, Tian, Dong and Liao86). Macrophages are the primary immune cells involved in kidney tissue, which also play a critical role in the cell–cell crosstalk in the kidney and development and progression of renal diseases (Ref. Reference Ebefors and Nyström85). Previous studies have demonstrated that tumour cell-derived lactate plays an important role in signal transduction by inducing tumour-associated macrophage polarisation and subsequently promoting tumour growth, which is also the case in renal cell carcinoma through the activation of specific receptors, such as GPR81 and GPR132, or through transport into cells by monocarboxylic acid transporters (Ref. Reference Colegio, Chu, Szabo, Chu, Rhebergen, Jairam, Cyrus, Brokowski, Eisenbarth, Phillips, Cline, Phillips and Medzhitov87). These findings also suggest that exploring the potential role of lactate in mediating the interactions among various cells in the local kidney throughout disease progression still holds considerable promise. In addition, the interaction between tubular epithelial cells and neighbouring endothelial cells is crucial for maintaining normal kidney function and plays a pivotal role in the pathogenesis and recovery of kidney diseases (Ref. Reference Tasnim and Zink88). However, the precise contribution of lactate in to this process remains unclear and warrants additional empirical investigation.

Signalling pathway

As a signalling molecule, lactate can activate various downstream signalling pathways by binding to specific receptors, including extracellular regulatory protein kinase (ERK1/2), nuclear factor κB (NF-κB) and Wnt signalling pathways, among others, thereby modulating the metabolic processes of tissues and cells (Refs. Reference Yang, Xu, Fan, Tu, Wang, Ha, Williams and Li89Reference Li, Wang, Wang, Chen and Liu91). This process is typically mediated through the cell surface receptor GPR81, which is independent of MCT, protons and cellular glucose metabolism and plays a crucial role in the progression of various systemic diseases, including tumours, cardiovascular diseases, brain diseases and more (Refs. Reference Li, Yang, Zhang, Lin, Fu, An, Zou, Wang, Wang and Yu17, Reference Meng92, Reference Ouyang, Wang and Huang93). GPR81 expression in the kidney has been investigated in previous studies using in situ hybridisation, revealing its predominant localisation at the vascular poles of the glomerulus and association with the renal microvascular system (Ref. Reference Wallenius, Thalén, Björkman, Johannesson, Wiseman, Böttcher, Fjellström and Oakes94). Through the utilisation of scRNA-seq techniques, Azushima’s team has additionally discovered that the renal expression of GPR81 is mainly localised mainly in mesangial cells (Ref. Reference Azushima72). When ischaemia–reperfusion occurs, extracellular lactate in the kidney is elevated, which can lead to GPR81 activation in mesangial cells that regulate the perfusion of large and vessels and microvessels in the kidney and reduce renal arterial blood flow, a process that relies on ET-1 signal transduction and ultimately exacerbates injury (Ref. Reference Jones, Stewart, Czopek, Menzies, Thomson, Moran, Cairns, Conway, Denby, Livingstone, Wiseman, Hadoke, Webb, Dhaun, Dear, Mullins and Bailey95). Furthermore, inflammatory studies have proposed that GPR81 acts as a mediator of the activation of macrophages and fibroblasts induced by lactate, contributing to renal interstitial inflammatory damage and fibrosis (Ref. Reference Manoharan, Prasad, Thangaraju and Manicassamy96). In addition, lactate can participate in signal transduction pathways in a nonreceptor-mediated manner. Lactate impedes renal protection by inhibiting autophagy initiation via the SIRT3/AMPK pathway in LPS-induced AKI, whereas inhibition of lactate production by 2-DG impedes apoptosis and enhances renal function (Ref. Reference Tan, Gu, Li, Chen, Liu, Liu, Zhang and Xiao67). Lactate-induced activation of the PD-1/PD-L1 pathway could suppress the immune response in SAKI models in mice (Ref. Reference Xu, Ma, Yu, Wang, Wang, Liu, Liu, Gao, Yu and Wang97). Additionally, lactate regulates the TRPV4/ TGFβ1/ SMAD2/3/CTGF signalling pathway to promote partial EMT and accelerate renal fibrosis in TECs (Ref. Reference Zhao, Xu, Chen, Cai, Gong, Li, Kuang, Liu, Zhou, Liu and Yin98).

However, to date, research on the signalling pathways associated with lactate activity in the kidney is still lacking, necessitating further exploration. Further research on signalling pathways involving lactate will facilitate a deeper understanding of its regulatory effects on various physiological and pathological processes.

Protein translational modification

Lactate can be converted to lactoyl-CoA, which is involved in the acylation of both histone and nonhistone proteins. By investigating core histones in human MCF7 cells, Zhang et al. has reported that the mass shift in the lysine residues of three proteolytic peptides is consistent with the addition of a lactyl group to lysine. This ground-breaking study provides the first evidence for the presence of lysine lactylation (Kla) and highlights Kla as a novel epigenetic modification in protein posttranslational processes (Ref. Reference Zhang, Tang, Huang, Zhou, Cui, Weng, Liu, Kim, Lee, Perez-Neut, Ding, Czyz, Hu, Ye, He, Zheng, Shuman, Dai, Ren, Roeder, Becker and Zhao99). The findings from this study suggest that lactate accumulated during glycolysis can serve as a precursor for lactate histone lysine, thereby directly influencing gene expression. Furthermore, Wan et al. then identified a cyclic ammonium ion, which serves as a reliable tool for the identification of novel Kla and modification sites, thereby revealing extensive modifications in the human proteome beyond histones (Ref. Reference Wan, Wang, Yu, Zhang, Tang, Wang, Lu, Li, Delafield, Kong, Wang, Shao, Lv, Wang, Tan, Wang, Hao and Ye100). Their findings demonstrated the presence of Kla proteins in the cytoplasm likewise and their predominant involvement in glycolysis. The presence of glycolytic inhibitors is directly correlated with lactate production and Kla, whereas mitochondrial inhibitors and cellular hypoxia can potentially increase lactate production and Kla activity (Refs. Reference Xiang, Zhao, Wu, Li, Fu and Ma101, Reference Shang, Liu and Hua102).

Protein translational modification (PTM) is a type of epigenetic modification. Through covalent modification, different acyl groups can attach to amino acid residues on histones or nonhistones and proteins undergo posttranslational changes in both the C-terminal region and the prominent N-terminal tail, thereby dynamically regulating protein localisation, activity and molecular interactions (Refs. Reference Xie, Hu, Liu, Zhou, Cheng, Huang and Cao103, Reference Millán-Zambrano, Burton, Bannister and Schneider104). Like other protein modifications, lactylation involves enzymes that catalyse the formation of specific types of PTMs (writers), as well as enzymes that remove PTMs (erasures) (Ref. Reference Millán-Zambrano, Burton, Bannister and Schneider104). P300, a well-known acetyltransferase that catalyses the acetylation of transcription factors, histones and other nucleoproteins and has been shown to be responsible for the induction of protein lactylation by lactate (Ref. Reference Zhang, Tang, Huang, Zhou, Cui, Weng, Liu, Kim, Lee, Perez-Neut, Ding, Czyz, Hu, Ye, He, Zheng, Shuman, Dai, Ren, Roeder, Becker and Zhao99). in vitro studies have shown that two deacetylase families, HDAC1–3 and SIRT1–3, reduce the expression of histones (including H3K18 and H4K5) Kla and that HDAC3 is the most effective erasing agent for Kla (Ref. Reference Moreno-Yruela, Zhang, Wei, Bæk, Liu, Gao, Danková, Nielsen, Bolding, Yang, Jameson, Wong, Olsen and Zhao105). In light of this, lactylation and acetylation are frequently coregulated. For instance, in a study conducted by Yang et al., lactate stimulated the lactylation and acetylation processes of high mobility group protein B1 in macrophages through the activation of p300 acetylase activity and inhibition of SIRT1 deacetylase activity (Ref. Reference Yang, Fan, Wang, Xu, Wang, Tu, Gill, Ha, Liu, Williams and Li106). These extensively investigated acetylation-related molecules in the context of kidney disease may also influence lactcylation and participate in the pathogenesis of this condition.

Since it was proposed, lactylation has been shown to be important for the functions of lactate, and it is involved in important life activities, such as glycolytic cell function, macrophage polarisation, vascular function, mitochondrial fission and fusion and nervous system regulation, including some recent findings in kidney disease. In exploring the pathogenesis of histone lactylation and clear cell renal cell carcinoma, Jiefeng Yang’s team reported a positive feedback loop between histone lactylation (H3K18la) and platelet-derived growth factor receptor β signalling, promoting tumour proliferation and migration (Ref. Reference Yang, Luo, Zhao, Li, Wang, Zeng, Yang, Zheng, Jie, Kang, Li, Liu, Zhou and Liu23). Previous studies conducted by our research team have revealed an increase in the level of glycosis in TECs following kidney injury, leading to the gradual accumulation of lactate, which subsequently increased the expression of the transcription factor Twist1 through lactylation at the histone H3K18 site, thereby promoting the progression of renal fibrosis. In addition, a new study revealed that lactate derived from the glycolytic reprogramming of renal tubules significantly enhances histone lactylation, specifically H4K12la, which is enriched on the promoters of NF-κB signal transduction genes and drives kidney fibrosis (Ref. Reference Wang107). Besides, the role of lactylation modification of nonhistone proteins in the occurrence and development of related kidney diseases has been gradually discovered. A study identified numerous increased lactylation sites on mitochondrial proteins that play key roles in mitochondrial metabolism in the kidneys of db/db mice via integrative lactylome analysis, and these results suggest that the lactylation of mitochondrial proteins may play a vital role in the progression of DKD, which warrants further investigation (Ref. Reference Chen, Feng, Qiao, Pan, Liang, Liu, Zhang, Liu, Liu and Liu108). Additionally, in patients with sepsis-induced acute kidney injury, lactate overproduction mediates mitochondrial fission protein 1 lysine 20 (Fis1 K20la) lactylation to promote excessive mitochondrial fission, thereby exacerbating kidney lesions and dysfunction (Ref. Reference An, Yao, Hu, Wu, Li, Li, Wu, Sun, Deng, Zhang, Gong, Huang, Chen and Zeng63). Lactylation of aldehyde dehydrogenase 2 at lysine 52 (ALDH2 K52la) promotes the ubiquitination-proteasomal degradation of prohibitin 2 (PHB2) – a crucial mitophagy receptor – thereby impairing mitophagy and exacerbating tubular injury and mitochondrial dysfunction in AKI (Ref. Reference Li, Shi, Xu, Wang, Hou, Luan and Chen109). Although few studies have investigated the association between lactylation and diseases in renal-related fields, we have shown that the increased production of lactate in the metabolic reprogramming of diseases such as AKI and DKD plays an important role in the progression of those diseases, and the potential impact of lactylation is also worthy of further exploration. Systematic analysis of the regulatory network between lactic acid metabolism and lactylation modification will help reveal their potential functional roles in the process of kidney injury and repair, thus opening up more possibilities for the treatment of diseases.

Prospects for clinical implementation

An in-depth study of the role of lactate metabolism in the physiological and pathological processes of the kidneys revealed that the dynamic changes in the levels of lactate and its related metabolic pathways potentially reflect the functional state of the kidneys. As mentioned earlier, existing research has shown that in diseases such as AKI and CKD, the processes of lactate generation, transport and clearance significantly change. For instance, the expression levels of MCTs in the kidneys are associated with renal function impairment. Current studies have demonstrated that elevated lactate levels in both serum and urine can serve as warning indicators for the occurrence and progression of AKI, DKD and other related renal disorders. In addition, MCT and LDH expression levels in renal tissue suggest disease progression and prognosis (Table 1). Therefore, the concentration of lactate or the expression levels of molecules related to its metabolism are expected to serve as biomarkers for the early diagnosis of kidney diseases, for assessing the degree of renal injury, predicting disease progression or monitoring treatment responses. Additionally, the integration of multiomics technologies (such as metabolomics and transcriptomics) holds promise for identifying more sensitive and specific combinations of lactate-related markers, providing more precise diagnostic tools for clinical use.

Table 1. The association between lactate and its metabolism-related molecules and the clinical progression of kidney diseases

Abbreviations: AKI, acute kidney injury; CysC, cystatin C; DKD, diabetic kidney disease; LDH, lactate dehydrogenase; MCT, monocarboxylate transporter; OS, overall survival; PFS, progression-free survival; PKM2, pyruvate kinase M2; RCC, renal cell carcinoma.

As research into the mechanistic roles of lactate in renal physiology and pathology progresses, therapeutic approaches targeting the lactate metabolic pathway are being increasingly recognised for their potential. Modulating the production, transport, or metabolic utilisation of lactate may represent a novel therapeutic strategy for the management of kidney diseases. First, regarding the generation of lactate, studies have shown that by treating diabetic mice with oxamate to inhibit LDHA or dichloroacetate to inhibit PDH phosphorylation, serum lactic acid levels are reversed, and renal function is simultaneously improved (Refs. Reference Zheng, Gu, Wang, Huang, Gou and Liu110, Reference Zhang, Chen, Lin, Huang, Wang, Xu, Wang, Chen, Zhang, Pan and Yin111). Furthermore, Xiang et al. demonstrated that shikonin can decrease lactate production in renal tubular cells through the inhibition of PKM2, resulting in reduced histone lactylation and alleviation of renal fibrosis in murine models (Ref. Reference Xiang, Wang, Huang, Zhou, Fei, Zhou, Yue, Li, Xue, Dai, Zhang, Ni, Sun and Huang112). Artesunate can reduce lactic acid levels by inhibiting glycolysis, thereby regulating the AMPK/mTOR pathway to suppress excessive autophagy and alleviate renal cell damage, improving sepsis-induced acute kidney injury (Ref. Reference Chen, du, Guo, Li, Zhang, Duan, Chen, Guo, Li and Jiang113). In addition, by regulating the expression or activity of MCTs, the distribution of lactic acid inside and outside cells can be altered, thereby promoting clearance and reducing the damage caused by the accumulation of lactic acid in renal tissue. Studies have shown that when syrosingopine, a dual inhibitor of the monocarboxylate transporters MCT1 and MCT4, is used to suppress lactic acid metabolic disorders, the development of renal tubular injury and fibrosis in uIRI mice is significantly reduced (Ref. Reference Tiwari, Sharma, Rajendran, Borkowski, An, Schonfeld, O’Sullivan, Schipma, Zhou, Courbon, Thomson, David, Quaggin, Thorp, Chandel and Kapitsinou114). However, current research remains confined to animal studies and lacks comprehensive evidence-based clinical evidence relevant to human medical practice. In recent years, drug development targeting lactate metabolism has made remarkable progress in the field of oncology, with several candidate drugs entering clinical trials, such as the MCT1 inhibitor AZD3965 and the anti-LDHA drug AT-101 (Refs. Reference Halford, Veal, Wedge, Payne, Bacon, Sloan, Dragoni, Heinzmann, Potter, Salisbury, Chénard-Poirier, Greystoke, Howell, Innes, Morris, Plummer, Rata, Petrides, Keun, Banerji and Plummer115, Reference Clem, O’Neal, Klarer, Telang and Chesney116). The potential of these lactate-related targeted drugs in metabolic regulation and pathological microenvironment remodelling also has biological relevance in kidney diseases, providing an important theoretical basis and research direction for their application in the treatment of kidney diseases.

Future investigations should incorporate clinical specimens, preclinical animal models and in vitro experimental systems to comprehensively elucidate the mechanistic role of lactate in both physiological kidney function and pathological processes while rigorously evaluating its translational potential in clinical settings. Such research endeavours will not only enhance the mechanistic understanding of kidney disease pathogenesis but also establish a solid theoretical foundation and empirical support for the development of novel diagnostic methodologies and therapeutic interventions.

Conclusion and outlook

Lactate is no longer just a metabolic waste product, we can see that it plays an important role in promoting or indicating disease progression in a variety of kidney diseases, through signal transduction or other means. However, at present, there is still a lack of systematic and clear research supporting for the specific mechanisms of lactate homeostasis in the kidneys under physiological conditions, especially the concept of the ‘lactate shuttle’. More importantly, in the pathological states of various kidney diseases, lactate homeostasis is often disrupted and the lactate concentration significantly increases. However, the specific change patterns, regulatory mechanisms and biological significance remain unclear. The role of lactate in kidney diseases cannot be simply categorised as either beneficial or detrimental and more in-depth research is needed. Therefore, in-depth research on the homeostatic regulation mechanism of lactic acid under normal physiological conditions in the kidneys, as well as its dynamic changes and functional significance at different pathological stages, helps to elucidate the complex role of lactic acid in the renal metabolic network.

In addition, by targeting the regulation of lactate production, and transport, and its associated molecular mechanism, innovative strategies for the future treatment of nephrological disorders may be developed. Furthermore, given that lactic acid metabolism involves multiple tissues and organs, systemic interventions may have broad physiological effects. Therefore, in the development of pharmacological agents or the formulation of therapeutic strategies, it is essential to incorporate targeted regulation of the kidneys to improve both the safety and efficacy of treatment. The intricate mechanism of action requires further exploration through clinical and basic research to substantiate its role and facilitate the development of pertinent strategies for clinical diagnosis and treatment.

Author contribution

K.G.: Writing—original draft, Writing—review and editing, Visualisation, Validation, Formal analysis, Data curation. Y.Z.: Writing—review and editing, Methodology, Investigation, Funding acquisition. S.G.: Writing—review and editing, Methodology. X.N.: Supervision, Resources, Funding acquisition, Conceptualisation. S.S.: Writing—review and editing, Supervision, Resources, Funding acquisition, Conceptualisation.

Funding statement

This study was supported by the National Natural Science Foundation of China grants (reference number: 82170722, 82270715) and National Natural Science Foundation for Young Scientists of China grants (reference number: 82300781).

Competing interests

The authors have no conflict of interest to declare.

Footnotes

K.G. and Y.Z. contributed equally to this work.

References

(2024) Kidney disease: A global health priority. Nature Reviews. Nephrology 20(7), 421423. https://doi.org/10.1038/s41581-024-00829-x.Google Scholar
Francis, A, Harhay, MN, Ong, ACM, Tummalapalli, SL, Ortiz, A, Fogo, AB, Fliser, D, Roy-Chaudhury, P, Fontana, M, Nangaku, M, Wanner, C, Malik, C, Hradsky, A, Adu, D, Bavanandan, S, Cusumano, A, Sola, L, Ulasi, I, Jha, V, American Society of Nephrology, European Renal Association and International Society of Nephrology (2024) Chronic kidney disease and the global public health agenda: An international consensus. Nature Reviews. Nephrology 20(7), 473485. https://doi.org/10.1038/s41581-024-00820-6.Google Scholar
Larcher, A, et al. (2025) Epidemiology of renal cancer: Incidence, mortality, survival, genetic predisposition, and risk factors. European Urology 88(4), 341358. https://doi.org/10.1016/j.eururo.2025.06.005.Google Scholar
Jefferis, J, Hudson, R, Lacaze, P, Bakshi, A, Hawley, C, Patel, C and Mallett, A (2024) Monogenic and polygenic concepts in chronic kidney disease (CKD). Journal of Nephrology 37(1), 721. https://doi.org/10.1007/s40620-023-01804-8.Google Scholar
Haas, R, Cucchi, D, Smith, J, Pucino, V, Macdougall, CE and Mauro, C (2016) Intermediates of metabolism: From bystanders to Signalling molecules. Trends in Biochemical Sciences 41(5), 460471. https://doi.org/10.1016/j.tibs.2016.02.003.Google Scholar
Brooks, GA (1985) Lactate:Glycolytic end product and oxidative substrate during sustained exercise in mammals – The “lactate shuttle”. Circulation, Respiration, and Metabolism. Berlin, Heidelberg: Springer Berlin Heidelberg, 208218.Google Scholar
Brooks, GA (2002) Lactate shuttles in nature. Biochemical Society Transactions 30(2), 258264. https://doi.org/10.1042/bst0300258.Google Scholar
Zhang, Y, Peng, Q, Zheng, J, Yang, Y, Zhang, X, Ma, A, Qin, Y, Qin, Z and Zheng, X (2023) The function and mechanism of lactate and lactylation in tumor metabolism and microenvironment. Genes Disease 10(5), 20292037. https://doi.org/10.1016/j.gendis.2022.10.006.Google Scholar
Certo, M, Tsai, CH, Pucino, V, Ho, PC and Mauro, C (2021) Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nature Reviews. Immunology 21(3), 151161. https://doi.org/10.1038/s41577-020-0406-2.Google Scholar
Chrysopoulou, M and Rinschen, MM (2024) Metabolic rewiring and communication: An integrative view of kidney proximal tubule function. Annual Review of Physiology 86, 405427. https://doi.org/10.1146/annurev-physiol-042222-024724.Google Scholar
van der Rijt, S, Leemans, JC, Florquin, S, Houtkooper, RH and Tammaro, A (2022) Immunometabolic rewiring of tubular epithelial cells in kidney disease. Nature Reviews. Nephrology 18(9), 588603. https://doi.org/10.1038/s41581-022-00592-x.Google Scholar
Miguel, V, Shaw, IW and Kramann, R (2025) Metabolism at the crossroads of inflammation and fibrosis in chronic kidney disease. Nature Reviews. Nephrology 21(1), 3956. https://doi.org/10.1038/s41581-024-00889-z.Google Scholar
Fan, X, Yang, M, Lang, Y, Lu, S, Kong, Z, Gao, Y, Shen, N, Zhang, D and Lv, Z (2024) Mitochondrial metabolic reprogramming in diabetic kidney disease. Cell Death & Disease 15(6), 442. https://doi.org/10.1038/s41419-024-06833-0.Google Scholar
Zhu, Z, Hu, J, Chen, Z, Feng, J, Yang, X, Liang, W and Ding, G (2022) Transition of acute kidney injury to chronic kidney disease: Role of metabolic reprogramming. Metabolism 131, 155194. https://doi.org/10.1016/j.metabol.2022.155194.Google Scholar
Shen, Y, Jiang, L, Wen, P, Ye, Y, Zhang, Y, Ding, H, Luo, J, Xu, L, Zen, K, Zhou, Y and Yang, J (2020) Tubule-derived lactate is required for fibroblast activation in acute kidney injury. American Journal of Physiology. Renal Physiology 318(3), F689F701. https://doi.org/10.1152/ajprenal.00229.2019.Google Scholar
Rabinowitz, JD and Enerback, S (2020) Lactate: The ugly duckling of energy metabolism. Nature Metabolism 2(7), 566571. https://doi.org/10.1038/s42255-020-0243-4.Google Scholar
Li, X, Yang, Y, Zhang, B, Lin, X, Fu, X, An, Y, Zou, Y, Wang, JX, Wang, Z, Yu, T (2022 ) Lactate Metabolism in Human Health and Disease. Signal Transduction and Targeted Therapy 7(1), 305. https://doi.org/10.1038/s41392-022-01151-3.Google Scholar
De Corte, W, et al. (2014) Severe lactic acidosis in critically ill patients with acute kidney injury treated with renal replacement therapy. Journal of Critical Care 29(4), 650655. https://doi.org/10.1016/j.jcrc.2014.02.019.Google Scholar
Bellomo, R (2002 ) Bench-to-bedside review: Lactate and the kidney. Critical Care 6(4), 322. https://doi.org/10.1186/cc1518.Google Scholar
Brooks, GA (2018) The science and translation of lactate shuttle theory. Cell Metabolism 27(4), 757785. https://doi.org/10.1016/j.cmet.2018.03.008.Google Scholar
Wu, D, Zhang, K, Khan, FA, Wu, Q, Pandupuspitasari, NS, Tang, Y, Guan, K, Sun, F and Huang, C (2023) The emerging era of lactate: A rising star in cellular signaling and its regulatory mechanisms. Journal of Cellular Biochemistry 124(8), 10671081. https://doi.org/10.1002/jcb.30458.Google Scholar
Frank, AC, Raue, R, Fuhrmann, DC, Sirait-Fischer, E, Reuse, C, Weigert, A, Lütjohann, D, Hiller, K, Syed, SN and Brüne, B (2021) Lactate dehydrogenase B regulates macrophage metabolism in the tumor microenvironment. Theranostics 11(15), 75707588. https://doi.org/10.7150/thno.58380.Google Scholar
Yang, J, Luo, L, Zhao, C, Li, X, Wang, Z, Zeng, Z, Yang, X, Zheng, X, Jie, H, Kang, L, Li, S, Liu, S, Zhou, C and Liu, H (2022) A positive feedback loop between inactive VHL-triggered histone Lactylation and PDGFRbeta signaling drives clear cell renal cell carcinoma progression. International Journal of Biological Sciences 18(8), 34703483. https://doi.org/10.7150/ijbs.73398.Google Scholar
Osis, G, Traylor, AM, Black, LM, Spangler, D, George, JF, Zarjou, A, Verlander, JW and Agarwal, A (2021) Expression of lactate dehydrogenase a and B isoforms in the mouse kidney. American Journal of Physiology. Renal Physiology 320(5), F706F718. https://doi.org/10.1152/ajprenal.00628.2020.Google Scholar
Wu, H, Malone, AF, Donnelly, EL, Kirita, Y, Uchimura, K, Ramakrishnan, SM, Gaut, JP and Humphreys, BD (2018) Single-cell transcriptomics of a human kidney allograft biopsy specimen defines a diverse inflammatory response. Journal of the American Society of Nephrology 29(8), 20692080. https://doi.org/10.1681/asn.2018020125.Google Scholar
Wu, H, Uchimura, K, Donnelly, EL, Kirita, Y, Morris, SA and Humphreys, BD (2018) Comparative analysis and refinement of human PSC-derived kidney organoid differentiation with single-cell transcriptomics. Cell Stem Cell 23(6), 869881.e8. https://doi.org/10.1016/j.stem.2018.10.010.Google Scholar
Zager, RA, Johnson, AC and Becker, K (2014) Renal cortical pyruvate depletion during AKI. Journal of the American Society of Nephrology 25(5), 9981012. https://doi.org/10.1681/asn.2013070791.Google Scholar
Zager, RA, Johnson, AC and Becker, K (2013) Renal cortical lactate dehydrogenase: A useful, accurate, quantitative marker of in vivo tubular injury and acute renal failure. PLoS One 8(6), e66776. https://doi.org/10.1371/journal.pone.0066776.Google Scholar
Jha, MK and Suk, K (2013) Pyruvate dehydrogenase kinase as a potential therapeutic target for malignant gliomas. Brain Tumor Research and Treatment 1(2), 5763. https://doi.org/10.14791/btrt.2013.1.2.57.Google Scholar
Jha, MK, Lee, IK and Suk, K (2016) Metabolic reprogramming by the pyruvate dehydrogenase kinase-lactic acid axis: Linking metabolism and diverse neuropathophysiologies. Neuroscience and Biobehavioral Reviews 68, 119. https://doi.org/10.1016/j.neubiorev.2016.05.006.Google Scholar
Luengo, A, Li, Z, Gui, DY, Sullivan, LB, Zagorulya, M, do, BT, Ferreira, R, Naamati, A, Ali, A, Lewis, CA, Thomas, CJ, Spranger, S, Matheson, NJ and Vander Heiden, MG (2021) Increased demand for NAD(+) relative to ATP drives aerobic glycolysis. Molecular Cell 81(4), 691707.e6. https://doi.org/10.1016/j.molcel.2020.12.012.Google Scholar
Kamel, KS, Oh, MS and Halperin, ML (2020) L-lactic acidosis: Pathophysiology, classification, and causes; emphasis on biochemical and metabolic basis. Kidney International 97(1), 7588. https://doi.org/10.1016/j.kint.2019.08.023.Google Scholar
Galgamuwa, R, Hardy, K, Dahlstrom, JE, Blackburn, AC, Wium, E, Rooke, M, Cappello, JY, Tummala, P, Patel, HR, Chuah, A, Tian, L, McMorrow, L, Board, PG and Theodoratos, A (2016) Dichloroacetate prevents cisplatin-induced nephrotoxicity without compromising cisplatin anticancer properties. Journal of the American Society of Nephrology 27(11), 33313344. https://doi.org/10.1681/asn.2015070827.Google Scholar
Lan, R, Geng, H, Singha, PK, Saikumar, P, Bottinger, EP, Weinberg, JM and Venkatachalam, MA (2016) Mitochondrial pathology and glycolytic shift during proximal tubule atrophy after ischemic AKI. Journal of the American Society of Nephrology 27(11), 33563367. https://doi.org/10.1681/asn.2015020177.Google Scholar
Yanase, H, Takebe, K, Nio-Kobayashi, J, Takahashi-Iwanaga, H and Iwanaga, T (2008) Cellular expression of a sodium-dependent monocarboxylate transporter (Slc5a8) and the MCT family in the mouse kidney. Histochemistry and Cell Biology 130(5), 957966. https://doi.org/10.1007/s00418-008-0490-z.Google Scholar
Halestrap, AP (2013) The SLC16 gene family - structure, role and regulation in health and disease. Molecular Aspects of Medicine 34(2–3), 337449. https://doi.org/10.1016/j.mam.2012.05.003.Google Scholar
Certo, M, Llibre, A, Lee, W and Mauro, C (2022) Understanding lactate sensing and signalling. Trends in Endocrinology and Metabolism 33(10), 722735. https://doi.org/10.1016/j.tem.2022.07.004.Google Scholar
Becker, HM, Mohebbi, N, Perna, A, Ganapathy, V, Capasso, G, and Wagner, CA (2010) Localization of members of MCT monocarboxylate transporter family Slc16 in the kidney and regulation during metabolic acidosis. American Journal of Physiology. Renal Physiology 299(1): F141F154. https://doi.org/10.1152/ajprenal.00488.2009.Google Scholar
Ippolito, L, Morandi, A, Giannoni, E and Chiarugi, P (2019) Lactate: A metabolic driver in the tumour landscape. Trends in Biochemical Sciences 44(2), 153166. https://doi.org/10.1016/j.tibs.2018.10.011.Google Scholar
Sheikh-Hamad, D (2021) Hints for a kidney lactate shuttle and lactomone. American Journal of Physiology. Renal Physiology 320(6), F1028F1029. https://doi.org/10.1152/ajprenal.00160.2021.Google Scholar
Iwanaga, T and Kishimoto, A (2015) Cellular distributions of monocarboxylate transporters: A review. Biomedical Research 36(5), 279301. https://doi.org/10.2220/biomedres.36.279.Google Scholar
Nakagawa, S, Nishihara, K, Miyata, H, Shinke, H, Tomita, E, Kajiwara, M, Matsubara, T, Iehara, N, Igarashi, Y, Yamada, H, Fukatsu, A, Yanagita, M, Matsubara, K and Masuda, S (2015) Molecular markers of tubulointerstitial fibrosis and tubular cell damage in patients with chronic kidney disease. PLoS One 10(8), e0136994. https://doi.org/10.1371/journal.pone.0136994.Google Scholar
Gopal, E, Umapathy, NS, Martin, PM, Ananth, S, Gnana-Prakasam, JP, Becker, H, Wagner, CA, Ganapathy, V and Prasad, PD (2007) Cloning and functional characterization of human SMCT2 (SLC5A12) and expression pattern of the transporter in kidney. Biochimica et Biophysica Acta 1768(11), 26902697. https://doi.org/10.1016/j.bbamem.2007.06.031.Google Scholar
Thangaraju, M, Ananth, S, Martin, PM, Roon, P, Smith, SB, Sterneck, E, Prasad, PD and Ganapathy, V (2006) C/ebpdelta null mouse as a model for the double knock-out of slc5a8 and slc5a12 in kidney. The Journal of Biological Chemistry 281(37), 2676926773. https://doi.org/10.1074/jbc.C600189200.Google Scholar
Legouis, D, Faivre, A, Cippà, PE and de Seigneux, S (2022) Renal gluconeogenesis: An underestimated role of the kidney in systemic glucose metabolism. Nephrology, Dialysis, Transplantation 37(8), 14171425. https://doi.org/10.1093/ndt/gfaa302.Google Scholar
Bankir, L (2021) Could an intrarenal Cori cycle participate in the urinary concentrating mechanism? American Journal of Physiology. Renal Physiology 321(3), F352f353. https://doi.org/10.1152/ajprenal.00253.2021.Google Scholar
Wuttke, M, et al. (2019) A catalog of genetic loci associated with kidney function from analyses of a million individuals. Nature Genetics 51(6), 957972. https://doi.org/10.1038/s41588-019-0407-x.Google Scholar
Liu, H, Abedini, A, Ha, E, Ma, Z, Sheng, X, Dumoulin, B, Qiu, C, Aranyi, T, Li, S, Dittrich, N, Chen, HC, Tao, R, Tarng, DC, Hsieh, FJ, Chen, SA, Yang, SF, Lee, MY, Kwok, PY, Wu, JY, Chen, CH, Khan, A, Limdi, NA, Wei, WQ, Walunas, TL, Karlson, EW, Kenny, EE, Luo, Y, Kottyan, L, Connolly, JJ, Jarvik, GP, Weng, C, Shang, N, Cole, JB, Mercader, JM, Mandla, R, Majarian, TD, Florez, JC, Haas, ME, Lotta, LA, Regeneron Genetics Center‡, GHS-RGC DiscovEHR Collaboration§, Drivas, TG, Penn Medicine BioBank¶, Vy, HMT, Nadkarni, GN, Wiley, LK, Wilson, MP, Gignoux, CR, Rasheed, H, Thomas, LF, Åsvold, BO, Brumpton, BM, Hallan, SI, Hveem, K, Zheng, J, Hellwege, JN, Zawistowski, M, Zöllner, S, Franceschini, N, Hu, H, Zhou, J, Kiryluk, K, Ritchie, MD, Palmer, M, Edwards, TL, Voight, BF, Hung, AM, Susztak, K, Baras, A, Abecasis, G, Ferrando, A, Coppola, G, Deubler, A, Economides, A, Lotta, LA, Overton, JD, Reid, JG, Shuldiner, A, Siminovitch, K, Portnoy, J, Jones, MB, Mitnaul, L, Fenney, A, Marchini, J, Revez Ferreira, MA, Ghoussaini, M, Nafde, M, Salerno, W, Overton, JD, Beechert, C, Fuller, E, Cremona, LM, Kalyuskin, E, du, H, Forsythe, C, Gu, Z, Guevara, K, Lattari, M, Lopez, A, Manoochehri, K, Challa, P, Pradhan, M, Reynoso, R, Schiavo, R, Padilla, MS, Wang, C, Wolf, SE, du, H, Guevara, K, Averitt, A, Banerjee, N, Li, D, Malhotra, S, Mower, J, Sarwar, M, Sharma, D, Yu, S, Zhang, A, Aqeel, M, Reid, JG, Nafde, M, Goyal, M, Mitra, G, Sreeram, S, Lanche, R, Mahajan, V, Vasireddy, SL, Eom, G, Punuru, KP, Gokhale, S, Sultan, B, Mule, P, Austin, E, Bai, X, Zhang, L, O’Keeffe, S, Panea, R, Edelstein, E, Rasool, A, Salerno, W, Maxwell, EK, Boutkov, B, Gorovits, A, Guan, J, Habegger, L, Hawes, A, Krasheninina, O, Zarate, S, Mansfield, AJ, Habegger, L, Abecasis, G, Backman, J, Burch, K, Campos, A, Ganel, L, Gaynor, S, Geraghty, B, Ghosh, A, Martinez, SR, Gillies, C, Gurski, L, Herman, J, Jorgenson, E, Joseph, T, Kessler, M, Kosmicki, J, Locke, A, Nakka, P, Marchini, J, Landheer, K, Delaneau, O, Ghoussaini, M, Marcketta, A, Mbatchou, J, Moscati, A, Pandey, A, Pandit, A, Ross, J, Sidore, C, Stahl, E, Thornton, T, Vedantam, S, Wang, R, Wu, KH, Ye, B, Zhang, B, Ziyatdinov, A, Zou, Y, Zhang, J, Watanabe, K, Tang, M, Wendt, F, Balasubramanian, S, Bao, S, Sun, K, Zhang, C, Ferrando, A, Coppola, G, Lotta, LA, Shuldiner, A, Siminovitch, K, Hobbs, B, Silver, J, Palmer, W, Guerreiro, R, Joshi, A, Baldassari, A, Willer, C, Graham, S, Mayerhofer, E, Castineira, EP, Haas, M, Verweij, N, Hindy, G, Bovijn, J, de, T, Akbari, P, Sun, L, Sosina, O, Gilly, A, Dornbos, P, Rodriguez-Flores, J, Riaz, M, Kapoor, M, Tzoneva, G, Jallow, MW, Alkelai, A, Ayer, A, Rajagopal, V, Gelfman, S, Kumar, V, Otto, J, Parikshak, N, Guvenek, A, Bras, J, Alvarez, S, Brown, J, He, J, Khiabanian, H, Revez, J, Skead, K, Zavala, V, Sul, JS, Chen, L, Choi, S, Damask, A, Lin, N, Paulding, C, Jones, MB, Chen, E, LeBlanc, MG, Mighty, J, Rico-Varela, J, Nishtala, N, Rana, N, Hernandez, J, Fenney, A, Schwartz, R, Hankins, J, Han, A, Hart, S, Perez-Beals, A, Solari, G, Rivera-Picart, J, Pagan, M, Siceron, S, Buchanan, A, Carey, DJ, Martin, CL, Meyer, M, Retterer, K, Rolston, D, Rader, DJ, Ritchie, MD, Weaver, JE, Naseer, N, Sirugo, G, Poindexter, A, Ko, YA, Nerz, KP, Livingstone, M, Vadivieso, F, DerOhannessian, S, Tran, T, Stephanowski, J, Santos, S, Haubein, N, Dunn, J, Verma, A, Morse Kripke, C, Risman, M, Judy, R, Wollack, C, Verma, SS, Damrauer, SM, Bradford, Y, Dudek, SM and Drivas, TG (2025) Kidney multiome-based genetic scorecard reveals convergent coding and regulatory variants. Science 387(6734), eadp4753. https://doi.org/10.1126/science.adp4753.Google Scholar
Liu, H, Doke, T, Guo, D, Sheng, X, Ma, Z, Park, J, Vy, HMT, Nadkarni, GN, Abedini, A, Miao, Z, Palmer, M, Voight, BF, Li, H, Brown, CD, Ritchie, MD, Shu, Y and Susztak, K (2022) Epigenomic and transcriptomic analyses define core cell types, genes and targetable mechanisms for kidney disease. Nature Genetics 54(7), 950962. https://doi.org/10.1038/s41588-022-01097-w.Google Scholar
Guo, T, Zhang, J, Wang, T, Yuan, Z, Tang, H, Zhang, D, Chen, S and Wang, X (2022) Lactic acid metabolism and transporter related three genes predict the prognosis of patients with clear cell renal cell carcinoma. Genes (Basel) 13(4), 620. https://doi.org/10.3390/genes13040620.Google Scholar
Sun, Z, Tao, W, Guo, X, Jing, C, Zhang, M, Wang, Z, Kong, F, Suo, N, Jiang, S and Wang, H (2022) Construction of a lactate-related prognostic signature for predicting prognosis, tumor microenvironment, and immune response in kidney renal clear cell carcinoma. Frontiers in Immunology 13, 818984. https://doi.org/10.3389/fimmu.2022.818984.Google Scholar
Wu, J, Wu, Y, Sun, Y, You, J, Zhang, W and Zhao, T (2025) Analysis of immune status and prognostic model incorporating lactate metabolism and immune-related genes in clear cell renal cell carcinoma. Discover Oncology 16(1), 1024. https://doi.org/10.1007/s12672-025-02746-2.Google Scholar
Sun, S, Li, H, Chen, J and Qian, Q (2017) Lactic acid: No longer an inert and end-product of glycolysis. Physiology (Bethesda) 32(6), 453463. https://doi.org/10.1152/physiol.00016.2017.Google Scholar
Madias, NE (1986) Lactic acidosis. Kidney International 29(3), 752764. https://doi.org/10.1038/ki.1986.62.Google Scholar
Wen, L, Li, Y, Li, S, Hu, X, Wei, Q and Dong, Z (2021) Glucose metabolism in acute kidney injury and kidney repair. Frontiers in Medicine (Lausanne) 8, 744122. https://doi.org/10.3389/fmed.2021.744122.Google Scholar
Legouis, D, Ricksten, SE, Faivre, A, Verissimo, T, Gariani, K, Verney, C, Galichon, P, Berchtold, L, Feraille, E, Fernandez, M, Placier, S, Koppitch, K, Hertig, A, Martin, PY, Naesens, M, Pugin, J, McMahon, AP, Cippà, PE and de Seigneux, S (2020) Altered proximal tubular cell glucose metabolism during acute kidney injury is associated with mortality. Nature Metabolism 2(8), 732743. https://doi.org/10.1038/s42255-020-0238-1.Google Scholar
García, AF, Manzano-Nunez, R, Bayona, JG, Naranjo, MP, Villa, DN, Moreno, M, Ossa, S, Martinez, JM, Martinez, N and Puyana, JC (2020) Acute kidney injury in severely injured patients admitted to the intensive care unit. Military Medical Research 7(1), 47. https://doi.org/10.1186/s40779-020-00277-1.Google Scholar
Zhang, Z and Ni, H (2015) Normalized lactate load is associated with development of acute kidney injury in patients who underwent cardiopulmonary bypass surgery. PLoS One 10(3), e0120466. https://doi.org/10.1371/journal.pone.0120466.Google Scholar
Harrois, A, et al. (2018) Prevalence and risk factors for acute kidney injury among trauma patients: A multicenter cohort study. Critical Care 22(1), 344. https://doi.org/10.1186/s13054-018-2265-9.Google Scholar
Choi, S, et al. (2024) High intraoperative serum lactate level is associated with acute kidney injury after brain tumor resection. Journal of Neurosurgical Anesthesiology 37(1), 5563. https://doi.org/10.1097/ana.0000000000000954.Google Scholar
Gómez, H, Kellum, JA and Ronco, C (2017) Metabolic reprogramming and tolerance during sepsis-induced AKI. Nature Reviews. Nephrology 13(3), 143151. https://doi.org/10.1038/nrneph.2016.186.Google Scholar
Xie, W, He, Q, Zhang, Y, Xu, X, Wen, P, Cao, H, Zhou, Y, Luo, J, Yang, J and Jiang, L (2023) Pyruvate kinase M2 regulates mitochondrial homeostasis in cisplatin-induced acute kidney injury. Cell Death & Disease 14(10), 663. https://doi.org/10.1038/s41419-023-06195-z.Google Scholar
An, S, Yao, Y, Hu, H, Wu, J, Li, J, Li, L, Wu, J, Sun, M, Deng, Z, Zhang, Y, Gong, S, Huang, Q, Chen, Z and Zeng, Z (2023) PDHA1 hyperacetylation-mediated lactate overproduction promotes sepsis-induced acute kidney injury via Fis1 lactylation. Cell Death & Disease 14(7), 457. https://doi.org/10.1038/s41419-023-05952-4.Google Scholar
Faivre, A, Verissimo, T, Auwerx, H, Legouis, D and de Seigneux, S (2021) Tubular cell glucose metabolism shift during acute and chronic injuries. Frontiers in Medicine (Lausanne) 8, 742072. https://doi.org/10.3389/fmed.2021.742072.Google Scholar
Comstock, J and Udenfriend, S (1970) Effect of lactate on collagen proline hydroxylase activity in cultured L-929 fibroblasts. Proceedings of the National Academy of Sciences of the United States of America 66(2): 552567. https://doi.org/10.1073/pnas.66.2.552.Google Scholar
Yang, Q, Huo, E, Cai, Y, Zhang, Z, Dong, C, Asara, JM and Wei, Q (2023) PFKFB3-mediated glycolysis boosts fibroblast activation and subsequent kidney fibrosis. Cells 12(16), 2081. https://doi.org/10.3390/cells12162081.Google Scholar
Tan, C, Gu, J, Li, T, Chen, H, Liu, K, Liu, M, Zhang, H and Xiao, X (2021) Inhibition of aerobic glycolysis alleviates sepsis-induced acute kidney injury by promoting lactate/sirtuin 3/AMPK-regulated autophagy. International Journal of Molecular Medicine 47(3), 19. https://doi.org/10.3892/ijmm.2021.4852.Google Scholar
Ding, H, Jiang, L, Xu, J, Bai, F, Zhou, Y, Yuan, Q, Luo, J, Zen, K and Yang, J (2017) Inhibiting aerobic glycolysis suppresses renal interstitial fibroblast activation and renal fibrosis. American Journal of Physiology. Renal Physiology 313(3), F561F575. https://doi.org/10.1152/ajprenal.00036.2017.Google Scholar
Verissimo, T, Faivre, A, Rinaldi, A, Lindenmeyer, M, Delitsikou, V, Veyrat-Durebex, C, Heckenmeyer, C, Fernandez, M, Berchtold, L, Dalga, D, Cohen, C, Naesens, M, Ricksten, SE, Martin, PY, Pugin, J, Merlier, F, Haupt, K, Rutkowski, JM, Moll, S, Cippà, PE, Legouis, D and de Seigneux, S (2022) Decreased renal gluconeogenesis is a Hallmark of chronic kidney disease. Journal of the American Society of Nephrology 33(4), 810827. https://doi.org/10.1681/asn.2021050680.Google Scholar
Sas, KM, Kayampilly, P, Byun, J, Nair, V, Hinder, LM, Hur, J, Zhang, H, Lin, C, Qi, NR, Michailidis, G, Groop, PH, Nelson, RG, Darshi, M, Sharma, K, Schelling, JR, Sedor, JR, Pop-Busui, R, Weinberg, JM, Soleimanpour, SA, Abcouwer, SF, Gardner, TW, Burant, CF, Feldman, EL, Kretzler, M, Brosius, FC III and Pennathur, S (2016) Tissue-specific metabolic reprogramming drives nutrient flux in diabetic complications. JCI Insight 1(15), e86976. https://doi.org/10.1172/jci.insight.86976.Google Scholar
Wu, Y, Dong, Y, Atefi, M, Liu, Y, Elshimali, Y and Vadgama, JV (2016) Lactate, a neglected factor for diabetes and cancer interaction. Mediators of Inflammation 2016, 6456018. https://doi.org/10.1155/2016/6456018.Google Scholar
Azushima, K. et al. (2023) Abnormal lactate metabolism is linked to albuminuria and kidney injury in diabetic nephropathy. Kidney International 104(6), 11351149. https://doi.org/10.1016/j.kint.2023.08.006.Google Scholar
Kanasaki, K (2023) The aberrant glycolysis in kidney proximal tubule: Potential therapeutic target for DKD. Kidney International 104(6), 10561059. https://doi.org/10.1016/j.kint.2023.09.019.Google Scholar
Srivastava, SP, Li, J, Kitada, M, Fujita, H, Yamada, Y, Goodwin, JE, Kanasaki, K and Koya, D (2018) SIRT3 deficiency leads to induction of abnormal glycolysis in diabetic kidney with fibrosis. Cell Death & Disease 9(10), 997. https://doi.org/10.1038/s41419-018-1057-0.Google Scholar
Song, C, Wang, S, Fu, Z, Chi, K, Geng, X, Liu, C, Cai, G, Chen, X, Wu, D and Hong, Q (2022) IGFBP5 promotes diabetic kidney disease progression by enhancing PFKFB3-mediated endothelial glycolysis. Cell Death & Disease 13(4), 340. https://doi.org/10.1038/s41419-022-04803-y.Google Scholar
Qi, W, Keenan, HA, Li, Q, Ishikado, A, Kannt, A, Sadowski, T, Yorek, MA, Wu, IH, Lockhart, S, Coppey, LJ, Pfenninger, A, Liew, CW, Qiang, G, Burkart, AM, Hastings, S, Pober, D, Cahill, C, Niewczas, MA, Israelsen, WJ, Tinsley, L, Stillman, IE, Amenta, PS, Feener, EP, Vander Heiden, MG, Stanton, RC and King, GL (2017) Pyruvate kinase M2 activation may protect against the progression of diabetic glomerular pathology and mitochondrial dysfunction. Nature Medicine 23(6), 753762. https://doi.org/10.1038/nm.4328.Google Scholar
Schlosser, P (2023) Advancing proteomics in nephrology: Unraveling causal pathways and therapeutic targets. Kidney International 104(6), 10591061. https://doi.org/10.1016/j.kint.2023.10.003.Google Scholar
Blantz, RC (2014) Phenotypic characteristics of diabetic kidney involvement. Kidney International 86(1), 79. https://doi.org/10.1038/ki.2013.552.Google Scholar
Chou, CK, Lee, YT, Chen, SM, Hsieh, CW, Huang, TC, Li, YC and Lee, JA (2015) Elevated urinary D-lactate levels in patients with diabetes and microalbuminuria. Journal of Pharmaceutical and Biomedical Analysis 116, 6570. https://doi.org/10.1016/j.jpba.2015.06.014.Google Scholar
Miranda-Gonçalves, V, Lameirinhas, A, Henrique, R, Baltazar, F and Jerónimo, C (2020) The metabolic landscape of urological cancers: New therapeutic perspectives. Cancer Letters 477, 7687. https://doi.org/10.1016/j.canlet.2020.02.034.Google Scholar
Miranda-Goncalves, V, et al. (2020) Lactate increases renal cell carcinoma aggressiveness through sirtuin 1-dependent epithelial mesenchymal transition Axis regulation. Cells 9(4), 1053. https://doi.org/10.3390/cells9041053.Google Scholar
Wei, G, et al. (2021) The thermogenic activity of adjacent adipocytes fuels the progression of ccRCC and compromises anti-tumor therapeutic efficacy. Cell Metabolism 33(10), e82039. https://doi.org/10.1016/j.cmet.2021.08.012.Google Scholar
Li, X, du, G, Li, L and Peng, K (2023) Cellular specificity of lactate metabolism and a novel lactate-related gene pair index for frontline treatment in clear cell renal cell carcinoma. Frontiers in Oncology 13, 1253783. https://doi.org/10.3389/fonc.2023.1253783.Google Scholar
Girgis, H, Masui, O, White, NMA, Scorilas, A, Rotondo, F, Seivwright, A, Gabril, M, Filter, ER, Girgis, AHA, Bjarnason, GA, Jewett, MAS, Evans, A, al-Haddad, S, Siu, KWM and Yousef, GM (2014) Lactate dehydrogenase a is a potential prognostic marker in clear cell renal cell carcinoma. Molecular Cancer 13, 101. https://doi.org/10.1186/1476-4598-13-101.Google Scholar
Ebefors, K and Nyström, J (2017) New insights into crosstalk in the kidney. Current Opinion in Nephrology and Hypertension 26(3), 143147. https://doi.org/10.1097/mnh.0000000000000310.Google Scholar
Guo, C, Cui, Y, Jiao, M, Yao, J, Zhao, J, Tian, Y, Dong, J and Liao, L (2023) Crosstalk between proximal tubular epithelial cells and other interstitial cells in tubulointerstitial fibrosis after renal injury. Frontiers in Endocrinology (Lausanne) 14, 1256375. https://doi.org/10.3389/fendo.2023.1256375.Google Scholar
Colegio, OR, Chu, NQ, Szabo, AL, Chu, T, Rhebergen, AM, Jairam, V, Cyrus, N, Brokowski, CE, Eisenbarth, SC, Phillips, GM, Cline, GW, Phillips, AJ and Medzhitov, R (2014) Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513(7519), 559563. https://doi.org/10.1038/nature13490.Google Scholar
Tasnim, F and Zink, D (2012) Cross talk between primary human renal tubular cells and endothelial cells in cocultures. American Journal of Physiology. Renal Physiology 302(8), F1055F1062. https://doi.org/10.1152/ajprenal.00621.2011.Google Scholar
Yang, K, Xu, J, Fan, M, Tu, F, Wang, X, Ha, T, Williams, DL and Li, C (2020) Lactate suppresses macrophage pro-inflammatory response to LPS stimulation by inhibition of YAP and NF-κB activation via GPR81-mediated signaling. Frontiers in Immunology 11, 587913. https://doi.org/10.3389/fimmu.2020.587913.Google Scholar
Madaan, A, Chaudhari, P, Nadeau-Vallée, M, Hamel, D, Zhu, T, Mitchell, G, Samuels, M, Pundir, S, Dabouz, R, Howe Cheng, CW, Mohammad Nezhady, MA, Joyal, JS, Rivera, JC and Chemtob, S (2019) Müller cell-localized G-protein-coupled receptor 81 (hydroxycarboxylic acid receptor 1) regulates inner retinal vasculature via norrin/Wnt pathways. The American Journal of Pathology 189(9), 18781896. https://doi.org/10.1016/j.ajpath.2019.05.016.Google Scholar
Li, G, Wang, HQ, Wang, LH, Chen, RP and Liu, JP (2014) Distinct pathways of ERK1/2 activation by hydroxy-carboxylic acid receptor-1. PLoS One 9(3), e93041. https://doi.org/10.1371/journal.pone.0093041.Google Scholar
Meng, X, et al. (2024) Lactate/hydroxycarboxylic acid receptor 1 in Alzheimer’s disease: Mechanisms and therapeutic implications-exercise perspective. Molecular Neurobiology 61(10), 77177731. https://doi.org/10.1007/s12035-024-04067-x.Google Scholar
Ouyang, J, Wang, H and Huang, J (2023) The role of lactate in cardiovascular diseases. Cell Communication and Signaling: CCS 21(1), 317. https://doi.org/10.1186/s12964-023-01350-7.Google Scholar
Wallenius, K, Thalén, P, Björkman, JA, Johannesson, P, Wiseman, J, Böttcher, G, Fjellström, O and Oakes, ND (2017) Involvement of the metabolic sensor GPR81 in cardiovascular control. JCI Insight 2(19), e92564. https://doi.org/10.1172/jci.insight.92564.Google Scholar
Jones, NK, Stewart, K, Czopek, A, Menzies, RI, Thomson, A, Moran, CM, Cairns, C, Conway, BR, Denby, L, Livingstone, DEW, Wiseman, J, Hadoke, PW, Webb, DJ, Dhaun, N, Dear, JW, Mullins, JJ and Bailey, MA (2020) Endothelin-1 mediates the systemic and renal hemodynamic effects of GPR81 activation. Hypertension 75(5), 12131222. https://doi.org/10.1161/hypertensionaha.119.14308.Google Scholar
Manoharan, I, Prasad, PD, Thangaraju, M and Manicassamy, S (2021) Lactate-dependent regulation of immune responses by dendritic cells and macrophages. Frontiers in Immunology 12, 691134. https://doi.org/10.3389/fimmu.2021.691134.Google Scholar
Xu, J, Ma, X, Yu, K, Wang, R, Wang, S, Liu, R, Liu, H, Gao, H, Yu, K and Wang, C (2021) Lactate up-regulates the expression of PD-L1 in kidney and causes immunosuppression in septic acute renal injury. Journal of Microbiology, Immunology, and Infection 54(3), 404410. https://doi.org/10.1016/j.jmii.2019.10.006.Google Scholar
Zhao, B, Xu, Y, Chen, Y, Cai, Y, Gong, Z, Li, D, Kuang, H, Liu, X, Zhou, H, Liu, G and Yin, Y (2022) Activation of TRPV4 by lactate as a critical mediator of renal fibrosis in spontaneously hypertensive rats after moderate- and high-intensity exercise. Frontiers in Physiology 13, 927078. https://doi.org/10.3389/fphys.2022.927078.Google Scholar
Zhang, D, Tang, Z, Huang, H, Zhou, G, Cui, C, Weng, Y, Liu, W, Kim, S, Lee, S, Perez-Neut, M, Ding, J, Czyz, D, Hu, R, Ye, Z, He, M, Zheng, YG, Shuman, HA, Dai, L, Ren, B, Roeder, RG, Becker, L and Zhao, Y (2019) Metabolic regulation of gene expression by histone lactylation. Nature 574(7779), 575580. https://doi.org/10.1038/s41586-019-1678-1.Google Scholar
Wan, N, Wang, N, Yu, S, Zhang, H, Tang, S, Wang, D, Lu, W, Li, H, Delafield, DG, Kong, Y, Wang, X, Shao, C, Lv, L, Wang, G, Tan, R, Wang, N, Hao, H and Ye, H (2022) Cyclic immonium ion of lactyllysine reveals widespread lactylation in the human proteome. Nature Methods 19(7), 854864. https://doi.org/10.1038/s41592-022-01523-1.Google Scholar
Xiang, T, Zhao, S, Wu, Y, Li, L, Fu, P and Ma, L (2022) Novel post-translational modifications in the kidneys for human health and diseases. Life Sciences 311(Pt B), 121188. https://doi.org/10.1016/j.lfs.2022.121188.Google Scholar
Shang, S, Liu, J and Hua, F (2022 ) Protein Acylation: Mechanisms, Biological Functions and Therapeutic Targets. Signal Transduction and Targeted Therapy 7(1), 396. https://doi.org/10.1038/s41392-022-01245-y.Google Scholar
Xie, Y, Hu, H, Liu, M, Zhou, T, Cheng, X, Huang, W and Cao, L (2022) The role and mechanism of histone lactylation in health and diseases. Frontiers in Genetics 13, 949252. https://doi.org/10.3389/fgene.2022.949252.Google Scholar
Millán-Zambrano, G, Burton, A, Bannister, AJ and Schneider, R (2022) Histone post-translational modifications - cause and consequence of genome function. Nature Reviews. Genetics 23(9), 563580. https://doi.org/10.1038/s41576-022-00468-7.Google Scholar
Moreno-Yruela, C, Zhang, D, Wei, W, Bæk, M, Liu, W, Gao, J, Danková, D, Nielsen, AL, Bolding, JE, Yang, L, Jameson, ST, Wong, J, Olsen, CA and Zhao, Y (2022) Class I histone deacetylases (HDAC1-3) are histone lysine delactylases. Science Advances 8(3), eabi6696. https://doi.org/10.1126/sciadv.abi6696.Google Scholar
Yang, K, Fan, M, Wang, X, Xu, J, Wang, Y, Tu, F, Gill, PS, Ha, T, Liu, L, Williams, DL and Li, C (2022) Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Cell Death and Differentiation 29(1), 133146. https://doi.org/10.1038/s41418-021-00841-9.Google Scholar
Wang, Y, et al. (2024) The glycolytic enzyme PFKFB3 drives kidney fibrosis through promoting histone lactylation-mediated NF-κB family activation. Kidney International 106(2), 226240. https://doi.org/10.1016/j.kint.2024.04.016.Google Scholar
Chen, J, Feng, Q, Qiao, Y, Pan, S, Liang, L, Liu, Y, Zhang, X, Liu, D, Liu, Z and Liu, Z (2024) ACSF2 and lysine lactylation contribute to renal tubule injury in diabetes. Diabetologia 67(7), 14291443. https://doi.org/10.1007/s00125-024-06156-x.Google Scholar
Li, J, Shi, X, Xu, J, Wang, K, Hou, F, Luan, X and Chen, L (2025) Aldehyde dehydrogenase 2 Lactylation aggravates mitochondrial dysfunction by disrupting PHB2 mediated mitophagy in acute kidney injury. Advancement of Science (Weinh) 12(8), e2411943. https://doi.org/10.1002/advs.202411943.Google Scholar
Zheng, T, Gu, YP, Wang, JM, Huang, TT, Gou, LS and Liu, YW (2025) Lactate-triggered histone lactylation contributes to podocyte epithelial-mesenchymal transition in diabetic nephropathy in mice. Chemico-Biological Interactions 408, 111418. https://doi.org/10.1016/j.cbi.2025.111418.Google Scholar
Zhang, X, Chen, J, Lin, R, Huang, Y, Wang, Z, Xu, S, Wang, L, Chen, F, Zhang, J, Pan, K and Yin, Z (2024) Lactate drives epithelial-mesenchymal transition in diabetic kidney disease via the H3K14la/KLF5 pathway. Redox Biology 75, 103246. https://doi.org/10.1016/j.redox.2024.103246.Google Scholar
Xiang, T, Wang, X, Huang, S, Zhou, K, Fei, S, Zhou, B, Yue, K, Li, Q, Xue, S, Dai, Y, Zhang, J, Ni, H, Sun, C and Huang, X (2025) Inhibition of PKM2 by shikonin impedes TGF-β1 expression by repressing histone lactylation to alleviate renal fibrosis. Phytomedicine 136, 156324. https://doi.org/10.1016/j.phymed.2024.156324.Google Scholar
Chen, H, du, P, Guo, R, Li, X, Zhang, L, Duan, J, Chen, M, Guo, J, Li, Y and Jiang, T (2025) Artesunate mitigates sepsis-induced acute kidney injury via lactate/AMPK/mTOR-regulated autophagy based on multi-omics. Drug Design, Development and Therapy 19, 59115928. https://doi.org/10.2147/dddt.S525701.Google Scholar
Tiwari, R, Sharma, R, Rajendran, G, Borkowski, GS, An, SY, Schonfeld, M, O’Sullivan, J, Schipma, MJ, Zhou, Y, Courbon, G, Thomson, BR, David, V, Quaggin, SE, Thorp, EB, Chandel, NS and Kapitsinou, PP (2024) Postischemic inactivation of HIF prolyl hydroxylases in endothelium promotes maladaptive kidney repair by inducing glycolysis. The Journal of Clinical Investigation 135(3), e176207. https://doi.org/10.1172/jci176207.Google Scholar
Halford, S, Veal, GJ, Wedge, SR, Payne, GS, Bacon, CM, Sloan, P, Dragoni, I, Heinzmann, K, Potter, S, Salisbury, BM, Chénard-Poirier, M, Greystoke, A, Howell, EC, Innes, WA, Morris, K, Plummer, C, Rata, M, Petrides, G, Keun, HC, Banerji, U and Plummer, R (2023) A phase I dose-escalation study of AZD3965, an Oral monocarboxylate transporter 1 inhibitor. Patients with Advanced Cancer Clinical Cancer Research 29(8), 14291439. https://doi.org/10.1158/1078-0432.Ccr-22-2263.Google Scholar
Clem, BF, O’Neal, J, Klarer, AC, Telang, S and Chesney, J (2016) Clinical development of cancer therapeutics that target metabolism. QJM 109(6), 367372. https://doi.org/10.1093/qjmed/hcv181.Google Scholar
Jiajun, W, Kaifeng, G and Jing, Z (2024) Urinary PKM2, a marker predicating acute kidney injury in patients with sepsis. International Urology and Nephrology 56(9), 30393045. https://doi.org/10.1007/s11255-024-04054-0.Google Scholar
Fang, Y, Zhang, Y, Shen, X, Dou, A, Xie, H, Zhang, Y and Xie, K (2025) Utilization of lactate trajectory models for predicting acute kidney injury and mortality in patients with hyperlactatemia: Insights across three independent cohorts. Renal Failure 47(1), 2474205. https://doi.org/10.1080/0886022x.2025.2474205.Google Scholar
Chen, M and Lv, D (2024) Prognostic value of serum lactate level for mortality in patients with acute kidney injury. European Journal of Medical Research 29(1), 295. https://doi.org/10.1186/s40001-024-01886-5.Google Scholar
Shi, X, Zhong, L, Lu, J, Hu, B, Shen, Q and Gao, P (2024) Clinical significance of the lactate-to-albumin ratio on prognosis in critically ill patients with acute kidney injury. Renal Failure 46(1), 2350238. https://doi.org/10.1080/0886022x.2024.2350238.Google Scholar
Liu, J, Min, J, Lu, J, Zhong, L and Luo, H (2024) Association between lactate/albumin ratio and prognosis in critically ill patients with acute kidney injury undergoing continuous renal replacement therapy. Renal Failure 46(2), 2374451. https://doi.org/10.1080/0886022x.2024.2374451.Google Scholar
Hou, Y, Deng, Y, Hu, L, He, L, Yao, F, Wang, Y, Deng, J, Xu, J, Wang, Y, Xu, F and Chen, C (2021) Assessment of 17 clinically available renal biomarkers to predict acute kidney injury in critically ill patients. Journal of Translational Internal Medicine 9(4), 273284. https://doi.org/10.2478/jtim-2021-0047.Google Scholar
Darshi, M, Kugathasan, L, Maity, S, Sridhar, VS, Fernandez, R, Limonte, CP, Grajeda, BI, Saliba, A, Zhang, G, Drel, VR, Kim, JJ, Montellano, R, Tumova, J, Montemayor, D, Wang, Z, Liu, JJ, Wang, J, Perkins, BA, Lytvyn, Y, Natarajan, L, Lim, SC, Feldman, H, Toto, R, Sedor, JR, Patel, J, Waikar, SS, Brown, J, Osman, Y, He, J, Chen, J, Reeves, WB, de Boer, IH, Roy, S, Vallon, V, Hallan, S, Gelfond, JA, Cherney, DZ, Sharma, K and Kidney Precision Medicine Project, and the CRIC Study Investigators (2024) Glycolytic lactate in diabetic kidney disease. JCI Insight 9(11), e168825. https://doi.org/10.1172/jci.insight.168825.Google Scholar
Lee, DY, Kim, JY, Ahn, E, Hyeon, JS, Kim, GH, Park, KJ, Jung, Y, LEE, YJ, Son, MK, Kim, SW, Han, SY, Kim, JH, Roh, GS, Cha, DR, Hwang, GS and Kim, WH (2022) Associations between local acidosis induced by renal LDHA and renal fibrosis and mitochondrial abnormalities in patients with diabetic kidney disease. Translational Research 249, 88109. https://doi.org/10.1016/j.trsl.2022.06.015.Google Scholar
Xiao, X, Zhang, J, Lang, Y, Cai, L, Yang, Q, Liu, K, Ji, S, Ju, X and Liu, F (2023) Associations of lactate dehydrogenase with risk of renal outcomes and cardiovascular mortality in individuals with diabetic kidney disease. Diabetes Research and Clinical Practice 203, 110838. https://doi.org/10.1016/j.diabres.2023.110838.Google Scholar
Tang, L, Yang, Q, Ma, R, Zhou, P, Peng, C, Xie, C, Liang, Q, Wu, T, Gao, W, Yu, H, Deng, G, Dai, Z, Mao, N and Xiao, X (2024) Association between lactate dehydrogenase and the risk of diabetic kidney disease in patients with type 2 diabetes. Frontiers in Endocrinology(Lausanne) 15, 1369968. https://doi.org/10.3389/fendo.2024.1369968.Google Scholar
Zhang, N, Zhang, H, Zhu, D, JiRiGaLa, , Yu, D, Wang, C, WuYunBiLiGe, , Amin, , ZhiHong, , Yu, H, Chen, X and Wang, M (2020) Prognostic role of pretreatment lactate dehydrogenase in patients with metastatic renal cell carcinoma: A systematic review and meta-analysis. International Journal of Surgery 79, 6673. https://doi.org/10.1016/j.ijsu.2020.05.019.Google Scholar
Ambrosetti, D, Dufies, M, Dadone, B, Durand, M, Borchiellini, D, Amiel, J, Pouyssegur, J, Rioux-Leclercq, N, Pages, G, Burel-Vandenbos, F and Mazure, NM (2018) The two glycolytic markers GLUT1 and MCT1 correlate with tumor grade and survival in clear-cell renal cell carcinoma. PLoS One 13(2), e0193477. https://doi.org/10.1371/journal.pone.0193477.Google Scholar
Cao, YW, Liu, Y, Dong, Z, Guo, L, Kang, EH, Wang, YH, Zhang, W and Niu, HT (2018) Monocarboxylate transporters MCT1 and MCT4 are independent prognostic biomarkers for the survival of patients with clear cell renal cell carcinoma and those receiving therapy targeting angiogenesis. Urologic Oncology 36(6), 311.e15311.e25. https://doi.org/10.1016/j.urolonc.2018.03.014.Google Scholar
Figure 0

Figure 1. Lactate transport and metabolism in the kidney under physiological and pathological circumstances. LDH and PDH coordinate the production and clearance of lactate, while MCTs facilitate lactate transport into or out of cells, collectively maintaining lactate homeostasis in the kidney. MCTs: monocarboxylate transporters; LDH: lactate dehydrogenase; PDH: pyruvate dehydrogenase; TCA: tricarboxylic acid. (Figure was created with Biorender.com.)

Figure 1

Figure 2. The expression of MCT1, MCT2 and MCT4 exhibit significant upregulation in the kidneys of patients with CKD. The mRNA levels of SLC16A1, SLC16A7 and SLC16A3 in kidney specimens of CKD (n = 53 samples) and control (n = 8 samples) in GSE66494 dataset are showed. Unpaired t-test, p < 0.05.

Figure 2

Figure 3. Schematic Diagram of the "lactate production-clearance axis" between the proximal and distal renal tubules. Under physiological conditions of the kidney, lactate is primarily generated in the proximal nephron under the catalysis of LDHA and is subsequently transported to the distal nephron via MCTs, where it is taken up by cells to meet their energy requirements or for signalling. (Figure was created with Biorender.com.)

Figure 3

Figure 4. The histopathological alterations in renal injury and fibrosis following lactate injection in the UIRI mouse model. LA: lactate intraperitoneal injection. Scale bar, 100 μm.

Figure 4

Figure 5. Lactate metabolism disorders in prevalent renal diseases and potential mechanisms of lactate’s involvement in those diseases’ progression. A. Elevated lactate concentration has been observed in the development of numerous renal diseases, yet its fluctuations and significance in the progression from AKI to CKD warrant further investigation. B. The occurrence of the above-mentioned diseases leads to alterations in the renal microenvironment due to increased lactate secretion. The diagram provides an overview of the potential mechanism by which lactate contributes to the progression of these diseases. Lactate is capable of directly facilitating the interaction between kidney cells, by activating specific signaling pathways, or entering cells for post-translational modifications to impact gene expression. GPR81: G-protein-coupled receptor 81; TRPV4: transient receptor potential vanilloid 4. MCT: monocarboxylate transporter; SIRT: sirtuin; HDAC: Histone deacetylase. (Figure was created with BioRender.com.)

Figure 5

Table 1. The association between lactate and its metabolism-related molecules and the clinical progression of kidney diseases