Skip to content
BY 4.0 license Open Access Published by De Gruyter Open Access September 11, 2023

Crosstalk between lactic acid and immune regulation and its value in the diagnosis and treatment of liver failure

  • Yong Lin ORCID logo , Gengjie Yan ORCID logo , Minggang Wang ORCID logo , Kan Zhang ORCID logo , Faming Shu ORCID logo , Meiyan Liu ORCID logo , Fuli Long ORCID logo EMAIL logo and Dewen Mao ORCID logo EMAIL logo
From the journal Open Life Sciences

Abstract

Liver failure is a common clinical syndrome of severe liver diseases, which belongs to one of the critical medical conditions. Immune response plays a leading role in the pathogenesis of liver failure. Lactic acid as a target for the treatment and prediction of liver failure has not attracted enough attention. Since the emergence of the concept of “histone lactation,” lactic acid has shown great promise in immune response and escape. Therefore, targeted lactic acid may be a reliable agent to solve immune and energy metabolism disorders in liver failure. Based on the relationship between lactic acid and immune response, the cross-talk between lactic acid metabolism, its compounds, and immune regulation and its significance in the diagnosis and treatment of liver failure were expounded in this article to provide new ideas for understanding and treating liver failure.

1 Introduction

Liver failure results from the continuous progression or sudden deterioration of various liver diseases. The high medical cost, mortality, and morbidity present a problematic situation of triple superposition, leading to liver failure becoming a major disease requiring comprehensive analysis of complex pathogenesis in the field of liver diseases. Based on the pathophysiological characteristics of liver failure, most studies still point to systemic immune inflammation playing a central role in the process of liver injury and determining the clinical outcome and prognosis [1]. Lactic acid is a metabolic product of pyruvate, which is the end product of glycolysis. Lactic acid not only provides energy for cell growth and development but also acts as an important signaling molecule affecting the biochemical functions of proteins in cells and regulates the biological functions of different kinds of cells [2,3]. Evidence shows that immune cells also consume a large amount of glucose during the immune response [4]. In addition, an immunosuppressive mechanism may be established in the presence of lactic acid to help inflammatory mediators and tumor cells obtain potential immune escape [5,6]. In this sense, lactic acid may act as an intermediary between immune response and immunosuppression [7]. Recently, Zhang et al. proposed a novel epigenetic change mediated by histone lysine lactoacylation [8]. Emerging studies have shown that histone lactation is involved in various cellular events, including immune regulation [2,3,4,8]. Therefore, lactic acid metabolism and its mechanism of action can help in understanding its association with the regulation of immune response during liver failure and act as an effective medium for liver disease diagnosis and treatment, which deserves our further attention.

2 Production and metabolism of lactic acid

In living organisms, cells use various metabolic pathways for energy generation and biosynthesis, of which glucose metabolism is the main process. Glucose is converted into pyruvate in the cytoplasm by glycolytic enzymes. At this time, pyruvate is at the crossroads of oxidative phosphorylation (OXPHOS) and fermentation, depending on the aerobic and anaerobic state of the cell [2]. Under aerobic conditions, pyruvate enters the tricarboxylic acid (TCA) cycle in the mitochondria to produce carbon dioxide and water. Each glucose molecule produces 36 adenosine triphosphate (ATP) molecules. In contrast, in the absence of oxygen, lactate dehydrogenase (LDH) reduces pyruvate from cytoplasm to lactic acid, producing only two ATPs [9]. LDH, pyruvate dehydrogenase kinase (PDK), and pyruvate dehydrogenase (PDH) control the conversion of pyruvate. LDH has two main subunits: LDHA and LDHB. Generally, LDHA catalyzes the conversion of pyruvate to lactic acid, while LDHB is responsible for the conversion of lactic acid to pyruvate [10]. The generated lactic acid continues to mediate the activity of PDK phosphorylation of PDH, leading to the obstruction of pyruvate entering the TCA cycle [11]. On the one hand, it reduces glucose consumption through OXPHOS; on the other hand, it causes pyruvate accumulation and indirectly promotes lactic acid production. In addition, the shuttle movement of lactic acid between cells and the microenvironment mainly depends on the monocarboxylic acid transporter (MCT) system, in which the inflow and outflow depend on MCT1 and MCT4, respectively [12]. When lactic acid is transferred between cells, it disrupts the pH homeostasis outside the cell, resulting in an acidic environment that affects enzyme activity and the regulation of immune cells. Consistent with this, energy metabolism preferentially switches from OXPHOS to glycolysis when the liver is damaged, resulting in the partial conversion of pyruvate to lactic acid [13]. From this perspective, the level of lactic acid is correlated with the degree of liver damage, and the production of lactic acid is closely related to the immune regulatory response of the liver.

3 Relationship between lactic acid and immune regulation in the liver

3.1 Nonspecific immunity

3.1.1 Dendritic cells (DCs)

DC is the strongest antigen-presenting cell in the human body, residing in the central vein and portal region of the liver. After antigen uptake, DC differentiates and matures, migrates to secondary lymphoid tissue, interacts with T cells initially, and initiates acquired immunity [14]. There are three subtypes of DCs in the liver: myeloid (MDCs), lymphoid (LDCs), and pre-plasma-cytoid DCs (pDCs). When the liver receives stimulation, glycolysis is enhanced, and a large amount of lactic acid is secreted within a few minutes after DC activation [15]. Generally, MDCs secrete IL-12 and induce Th1 cells and cytotoxic T lymphocyte (CTL) immune response, but studies have found that the increase in exogenous lactic acid can inhibit the differentiation and maturation of MDCs [16]. LDCs induce a Th2 immune response. When stimulated by foreign antigens, LDCs and pDCs produce a large amount of type I interferons, which directly inhibit viral replication and activate natural killer (NK) cells, B cells, T cells, and MDCs to induce and enhance antiviral immune response. Lactate can interact with the lactate receptor G-protein-coupled receptor 81 on pDCs to inhibit antigen presentation by interfering with antigen degradation in the endosomes [17]. In addition, lactate enhances tryptophan metabolism and kynurenine production by pDCs, which contribute to the induction of FoxP3 CD4 Tregs [18]. Furthermore, the liver immunomodulatory analysis showed that during liver inflammation or in solid tumors of the liver, the continuous increase in lactic acid transformation results in high lactate levels in cells, while DCs in an acidic environment are characterized by reduced glucose consumption, increased lactate output, and upregulated mitochondrial oxidative metabolism [19], and the combined action of the two inevitably interferes with immune initiation and response.

3.1.2 NK and natural killer T (NKT) cells

In the liver, the NK cells, unlike T and B cells, can directly produce a target cell-killing effect without specific antigen stimulation and secrete many inflammatory factors, such as IFN-γ, TNF-α, and IL-3, which play a vital role in antiviral and immune regulation of the body [20]. NK cells can be divided into two types based on the expression of transcription factors, namely circulating conventional NK cells in the blood and tissue-resident NK cells (trNK) [21]. Studies have shown that LDHA-mediated aerobic glycolysis and OXPHOS are essential for NK cell proliferation and maintenance of its antiviral and antitumor capabilities [22,23]. For example, in tumors or virus-carrying liver, impaired mitochondrial function mediated by elevated lactate levels leads to early apoptosis of trNK cells [21]. However, in the cellular microenvironment, the decrease in the pH value of tumor cells further amplifies the lactic acid-inhibited immune response [24] because the tumor-derived lactic acid downregulates the expression of the activated NK receptors NKp46 and NKG2D. This results in the decreased expression of perforin and granase [25,26], thus reducing the antiviral activity of NK cells. Conversely, blocking the flow of lactic acid into NK cells or increasing the oxygen level in the microenvironment can restore or enhance the toxic function of NK cells and cytokine production [27,28]. NKT cells originate from the thymus, express receptors on the surface of T cells and NK cells, and are activated by recognizing the antigen presented by the MHC molecule CD1d to release several cytokines such as IFN-γ and IL-4, regulate the balance between Th1 and Th2, and participate in important processes such as immune diseases and antitumor and antiviral activities in the liver. Studies have shown that NKT cells depend more on OXPHOS to proliferate and promote cytokine expression. However, high levels of extracellular lactic acid block glycolysis in NKT cells, negatively affecting their survival and functional expression [29].

3.1.3 Macrophage

In liver immunity, macrophages mainly express surface molecules related to antigen uptake, including complement receptors, scavenger receptors, and toll-like receptors, and take up exogenous antigens and present them to activated effector T cells, enhancing T cell biological activity. Macrophage polarization can be mainly divided into two types: classically activated macrophages (M1 macrophages) and alternately activated macrophages (M2 macrophages) [30]. M1 macrophages are highly effective effector cells with anti-inflammatory and immune regulation effects, while M2 macrophages are involved in pro-inflammatory processes, adaptive Th1 immunity, tissue remodeling and repair, and tumor progression [31]. Studies have shown that LDHA plays a key role in regulating macrophage polarization. The loss of LDHA in macrophages promotes polarization of M1-like macrophages, resulting in decreased expression of vascular endothelial growth factor (VEGF) and increased activity of effector CD8+ T cells [32]. On the contrary, LDHA promotes the polarization of M2 macrophages by promoting the expression of hypoxia-inducing factor HIF-1a, increasing the expression of VEGF [33]. So far, macrophages have been found to facilitate the M1-to-M2 and M2-to-M1 phenotype transitions. However, a recent study showed that tumor acidosis induces the phenotypic transformation of regulatory macrophages, which promotes tumor growth. The specific mechanism involves the activation of plasma membrane G-protein-coupled receptors mediated by acidosis and downstream cyclic adenosine phosphate signaling. This blocks the M2 gene transcription and thus inhibits the expression of inflammatory genes TNF and Nos2 [34], suggesting that lactic acid may mediate partial cross-talk between tumor cells and macrophages. From similar conclusions, it can be inferred that lactic acid promotes the expression of inflammatory bodies and pro-inflammatory factors such as IL-1β, IL-10, IL-6, and HIF-1α by downregulating the phosphorylation of p65-NFκB in macrophages or enhancing TLR4 signaling in macrophages [35,36]. Although the specific cross-talk mechanism between lactic acid and macrophages is still unclear, it is certain that long-term exposure of cells to lactic acid leads to mitochondrial autophagy in macrophages, causing effects such as decreased mitochondrial function, increased reactive oxygen species, and impaired oxidative ATP production, which damage the function of macrophages [37].

3.2 Specific immunity

3.2.1 CD4+ T cells

CD4+ T cells include Th1, Th2, CD4+ Treg, and Th17 cells. When T cells are activated, aerobic glycolysis converts glucose into lactic acid to meet energy and biosynthesis requirements [38]. Evidence has shown that lactic acid is directly or indirectly involved in CD4+ T cell expression. Intuitively, lactic acid regulates CD4+ T cell polarization and reduces the percentage of anti-tumor Th1 subpopulation by inducing silencing regulatory protein silent information regulator sirtuin 1 (SIRT1)-mediated T-box expressed in T cells (T-bet) transcription factor deacetylation [39]. Indirectly, LDHA is a key enzyme converting pyruvate into lactic acid. It regulates the expression of IFN-γ in Th1 cells by regulating the acetylation of histone H3 at lysine 9 and lysine 3 (H3K3me3) (Th1 cells are mainly involved in the immune response by secreting IL-2 and IFN-γ) [40]. In addition, lactic acid directly inhibits the movement of T cells in inflammatory tissues, leading to the trapping of T cells and the production of pro-inflammatory cytokines to amplify immune inflammatory response [41]. This may be attributed to the fact that when the extracellular lactic acid level is high, lactic acid enters CD4+ T cells through MCT1 and converts into pyruvate through LDHB, resulting in the downregulation of T cell glycolysis, blocking the output of lactic acid by T cells, and the accumulated lactic acid disrupts CTL metabolism [42]. Similarly, a lactate-rich environment interferes with the glycolysis of CD4+ T cells through the lactate transporter SLC5A12 and reduces their chemotaxis to the chemokine CXCL10, thus preferentially differentiating Th17 cells and inflammatory subsets [43].

The major subtype of Treg, CD4+ CD25+ Treg, is a group of T cells that specifically express the transcription factor Foxp3, and its ratio to effector T cells plays an important role in immunosuppression and maintenance of immune balance. The lactic acid in the tumor microenvironment helps Tregs to proliferate and maintain their immunosuppressive function [44]. This can be because lactic acid is transported to the initial CD4+ T cells via MCT1, which activates the expression of NF-κB and Foxp3 (NF-κB is a key regulatory factor of Foxp3 transcription). The high expression of Foxp3 upregulates the proportion of Tregs by inhibiting glycolysis and enhancing OXPHOS, leading to the decline in anti-tumor immunity. On the contrary, high glucose or low lactate levels can inhibit its function and stability [45]. In addition, Tregs use lactic acid to promote the entry of the nuclear factor of activated T cell 1 into the nucleus and induce the expression of programmed death receptor 1, thus making Treg cells more adaptable to low glucose and high lactic acid levels [46].

3.2.2 CD8+ T cells

CD8+ T cells primarily recognize antigen polypeptides presented by MHC Class I molecules. Similar to CD4+ T cells, LDHA deficiency reduces the antitumor activity of CD8+ T cells and may also prevent their movement and proliferation [47]. High levels of glycolytic enzymes are also critical for CD8+ T cells to express IFN-γ during an immune response [48]. In contrast to the T-reg response, lactic acid from tumor cells inhibits NFAT expression in CD8+ T cells, thus reducing the production of IFN-γ and maintaining immune balance with T-reg cells. However, excess lactic acid in tumor cells leads to immune tolerance, and neutralization with proton pump inhibitors can restore the T cell function [49]. The specific mechanism may be that a high lactate level damages the c-Jun N-terminal kinase pathway and the phosphorylation of p38 protein triggered by T cell receptors, thus inhibiting the function of CTL [50] (Figure 1).

Figure 1 
                     Effect of lactic acid on immune cells in an inflammatory microenvironment. Lactate induces immunosuppression, acidosis, and immune escape by regulating the expression of various genes.
Figure 1

Effect of lactic acid on immune cells in an inflammatory microenvironment. Lactate induces immunosuppression, acidosis, and immune escape by regulating the expression of various genes.

4 Lactic acid is closely related to liver failure

Acute injury during liver failure leads to the activation of innate immune cells, triggering a cascade of cytokines and chemokines, followed by an aggressive systemic inflammatory response syndrome, which is the overall characteristic of immune disorders in liver failure. There are many interactions between host innate immune activation and adaptive immune response to control the overall immune outcome of liver failure. From the above discussion, we know that lactic acid synthesis and secretion are embodied in the whole process of the immune response during liver failure (Figure 2). The effect of lactate levels on liver failure was described in the Lancet in 2002 [51]. Cohort studies have shown that arterial lactate levels are significantly higher in patients with acute liver failure (ALF), and their levels are more rapid and accurate in identifying the outcome of patients with paracetamol-induced ALF [51]. Subsequent studies have been more broadly interpreted [52,53,54,55]. For example, early lactate value is a strong marker of post-hepatectomy liver failure and has the potential to guide postoperative care [52]. The combination of NK cell frequency and lactate levels on admission can reliably predict the survival rate of ALF patients [53]. However, a cohort study found that the LDH level had a prognostic value similar to the end-stage liver disease model (MELD), and the combined prediction method was superior to the two considered separately [54]. Interestingly, the combination of lactic acid levels with the chronic liver failure-sequential organ failure assessment score (CLIF-SOFA) for sequential organ failure also significantly improved the prediction of short-term prognosis in patients with HBV-acute-on-chronic liver failure compared with the use of only CLIF-SOFA [55]. Moreover, during severe complications associated with liver failure (ketoacidosis, hepatic encephalopathy, and sepsis), lactic acid elevation can be an independent risk factor [56,57,58]. Furthermore, lactic acid prediction is applicable in hepatic encephalopathy and high intracranial pressure associated with liver failure. An elevated lactate/pyruvate ratio indicates that the accumulation of glutamine impairs mitochondrial function and leads to intracranial hypertension [59], and lactate is involved in the physiological and pathological reactions of liver failure from the early stage [60]. In contrast, pyruvate kinase deficiency can lead to severe liver dysfunction [61]. In addition, enzymes and compounds related to lactate synthesis and metabolism have shown similar values in animal research as clinical findings. The translocation of PDH and LDH to the nucleus of liver failure mice resulted in increased nuclear concentrations of acetyl-CoA and lactic acid and led to the expression of histone H3 hyperacetylation and damage response genes. Inhibitors of the two enzymes can reduce liver injury and improve survival rates [62]. Ethyl pyruvate (EP) can reduce intestinal permeability, inhibit a variety of pro-inflammatory cytokines in ALF rats, and protect rat liver function [63]. Recently, Zhou et al. [64] determined that contrary to their typical role as anti-inflammatory agents in the host, indole-3-acetic acid (IAA) and indole-3-lactic acid (ILA) gavage-sensitized mice to D-GalN/LPS-induced-ALF with a rapid increase in serum transaminases and histologic lesion. This can be attributed to the exacerbation of D-GalN/LPS-induced ALF via the probable involvement of the Tlr2/NF-κB pathway and ileac dysbiosis characterized by enriched Gram-positive genus due to the IAA pretreatment. In addition, Gan et al. [65] found that the acidic microenvironment caused by elevated lactic acid could inhibit the production and function of CD25, CD3, Foxp3, and iTregs through the PI4K/Akt/mTOR signaling pathway, leading to liver dysfunction. In contrast, the reversal of the acidic microenvironment restored Foxp3 expression and iTreg function. Moreover, the proton pump inhibitor omeprazole improved the decreased iTreg differentiation caused by the acidic microenvironment.

Figure 2 
               Diagram showing crosstalk between lactic acid and immune regulation. The crosstalk mechanism between lactic acid and various immune cells is shown in the box.
Figure 2

Diagram showing crosstalk between lactic acid and immune regulation. The crosstalk mechanism between lactic acid and various immune cells is shown in the box.

5 Summary

The physiological and pathological characteristics of lactic acid play a wide range of roles in immune regulatory responses. It is certain that elevated lactate levels not only inhibit the anti-inflammatory and anti-tumor effects of immune cells such as CD4+ T cells, CD8+ T cells, NK cells, and NKT cells but also benefit immunosuppressive cells such as Treg cells. The physiological vector response of lactic acid can maintain the proportion of effector T cells and maintain immune balance. Pathologically, with the increase in lactic acid levels, the energy metabolism cycle, mainly glycolysis, is seriously affected, and the immune balance is immediately destroyed, and even immune escape and immunosuppression occur. During liver failure, the metabolism of lactic acid may be closely related to the continuous disorder and sudden outbreak of immune inflammatory response, and the diagnosis and treatment of liver failure targeting lactic acid, such as LDHA and EP, has shown great developmental prospects. However, lactic acid as a therapeutic strategy still faces many challenges. For example, the starting point of lactic acid and immune regulation is the relationship between immune cells and glucose metabolites; whether this relationship involves energy supply, redox action, glucose conversion, glycolytic metabolism, and many other physiological links is yet to be explored. At the same time, it is worth noting that lactic acid and immune cells are also associated with several low molecular compounds, such as caffeine [66], solute carrier transporters [67], and tryptophan (including ILA, indole-3-acrylate, and indole-3-propionic acid) [68]. Therefore, the relation between liver failure and other liver diseases and lactic acid and lactate needs to be further explored, and this concept can help improve the diagnosis and treatment strategy for liver failure in the future.

Acknowledgments

The author would like to thank Professors Mao Dewen, Long Fuli, and Wang Minggang for their useful discussions on topics related to this work. This work has received funding support from the Guangxi Science and Technology Project of the National Natural Science Foundation of China (82260907, 82260899, 82274434) (2020GXNSFAA297098, 2020GXNSFAA297205, 2020GXNSFAA297206, AB22035076, 2020GXNSFAA297070). Finally, I would like to thank Xing Lin for her assistance in the digital compilation of this paper.

  1. Funding information: This study was financially supported by the Guangxi Natural Science Foundation (nos. 2020GXNSFAA297098, 2020GXNSFAA297205, 2020GXNSFAA297206, AB22035076, 2020GXNSFAA297070) and National Natural Science Foundation of China (nos. 82260907, 82260899, 82274434).

  2. Author contributions: Conceptualization: Yong Lin, Dewen Mao. Methodology: Yong Lin, Genjie Yan, Fuli Long. Data Curation: Kan Zhang, Faming Shu, Meiyan Liu. Analysis and/or Interpretation: Yong Lin, Gengjie Yan. Supervision: YongLin, Gengjie Yan, Minggang Wang. Writing - Original Draft: Yong Lin. Writing - Review & Editing: Yong Lin, Dewen Mao, Fuli Long.

  3. Conflict of interest: Authors state no conflict of interest.

  4. Data availability statement: Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

[1] Puengel T, Tacke F. Repair macrophages in acute liver failure. Gut. 2018 Feb;67(2):202–3.10.1136/gutjnl-2017-314245Search in Google Scholar PubMed

[2] Ye L, Jiang Y, Zhang M. Crosstalk between glucose metabolism, lactate production and immune response modulation. Cytokine Growth Factor Rev. 2022 Dec;68:81–92.10.1016/j.cytogfr.2022.11.001Search in Google Scholar PubMed

[3] Zhang Y, Zhai Z, Duan J, Wang X, Zhong J, Wu L, et al. Lactate: The Mediator of Metabolism and Immunosuppression. Front Endocrinol (Lausanne). 2022 Jun 9;13:901495.10.3389/fendo.2022.901495Search in Google Scholar PubMed PubMed Central

[4] Reinfeld BI, Madden MZ, Wolf MM, Chytil A, Bader JE, Patterson AR, et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature. 2021 May;593(7858):282–8.10.1038/s41586-021-03442-1Search in Google Scholar PubMed PubMed Central

[5] Neagu M. Metabolic Traits in Cutaneous Melanoma. Front Oncol. 2020 May 19;10:851.10.3389/fonc.2020.00851Search in Google Scholar PubMed PubMed Central

[6] Ganapathy-Kanniappan S. Linking tumor glycolysis and immune evasion in cancer: Emerging concepts and therapeutic opportunities. Biochim Biophys Acta Rev Cancer. 2017 Aug;1868(1):212–20.10.1016/j.bbcan.2017.04.002Search in Google Scholar PubMed

[7] Kelly B, O'Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015 Jul;25(7):771–84.10.1038/cr.2015.68Search in Google Scholar PubMed PubMed Central

[8] Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019 Oct;574(7779):575–80.10.1038/s41586-019-1678-1Search in Google Scholar PubMed PubMed Central

[9] Tu VY, Ayari A, O'Connor RS. Beyond the lactate paradox: How lactate and acidity impact T cell therapies against cancer. Antibodies (Basel). 2021 Jun 28;10(3):25.10.3390/antib10030025Search in Google Scholar PubMed PubMed Central

[10] Lee TY. Lactate: a multifunctional signaling molecule. Yeungnam Univ J Med. 2021 Jul;38(3):183–93.10.12701/yujm.2020.00892Search in Google Scholar PubMed PubMed Central

[11] Hong SM, Lee YK, Park I, Kwon SM, Min S, Yoon G. Lactic acidosis caused by repressed lactate dehydrogenase subunit B expression down-regulates mitochondrial oxidative phosphorylation via the pyruvate dehydrogenase (PDH)-PDH kinase axis. J Biol Chem. 2019 May 10;294(19):7810–20.10.1074/jbc.RA118.006095Search in Google Scholar PubMed PubMed Central

[12] Zhao Y, Li W, Li M, Hu Y, Zhang H, Song G, et al. Targeted inhibition of MCT4 disrupts intracellular pH homeostasis and confers self-regulated apoptosis on hepatocellular carcinoma. Exp Cell Res. 2019 Nov 1;384(1):111591.10.1016/j.yexcr.2019.111591Search in Google Scholar PubMed

[13] Nishikawa T, Bellance N, Damm A, et al. A switch in the source of ATP production and a loss in capacity to perform glycolysis are hallmarks of hepatocyte failure in advance liver disease. J Hepatol. 2014 Jun;60(6):1203–11.10.1016/j.jhep.2014.02.014Search in Google Scholar PubMed PubMed Central

[14] Patente TA, Pelgrom LR, Everts B. Dendritic cells are what they eat: how their metabolism shapes T helper cell polarization. Curr Opin Immunol. 2019 Jun;58:16–23.10.1016/j.coi.2019.02.003Search in Google Scholar PubMed

[15] Tiberio L, Del Prete A, Schioppa T, Sozio F, Bosisio D, Sozzani S. Chemokine and chemotactic signals in dendritic cell migration. Cell Mol Immunol. 2018 Apr;15(4):346–52.10.1038/s41423-018-0005-3Search in Google Scholar PubMed PubMed Central

[16] Marin E, Bouchet-Delbos L, Renoult O, Louvet C, Nerriere-Daguin V, Managh AJ, et al. Human tolerogenic dendritic cells regulate immune responses through lactate synthesis. Cell Metab. 2019 Dec 3;30(6):1075–90.e8.10.1016/j.cmet.2019.11.011Search in Google Scholar PubMed

[17] Selleri S, Bifsha P, Civini S, Pacelli C, Dieng MM, Lemieux W, et al. Human mesenchymal stromal cell-secreted lactate induces M2-macrophage differentiation by metabolic reprogramming. Oncotarget. 2016 May 24;7(21):30193–210.10.18632/oncotarget.8623Search in Google Scholar PubMed PubMed Central

[18] Raychaudhuri D, Bhattacharya R, Sinha BP, Liu CSC, Ghosh AR, Rahaman O, et al. Lactate induces pro-tumor reprogramming in intratumoral plasmacytoid dendritic cells. Front Immunol. 2019 Aug 7;10:1878.10.3389/fimmu.2019.01878Search in Google Scholar PubMed PubMed Central

[19] Erra Díaz F, Ochoa V, Merlotti A, Dantas E, Mazzitelli I, Gonzalez Polo V, et al. Extracellular acidosis and mTOR inhibition drive the differentiation of human monocyte-derived dendritic cells. Cell Rep. 2020 May 5;31(5):107613.10.1016/j.celrep.2020.107613Search in Google Scholar PubMed

[20] Wang J, Liu X, Jin T, Cao Y, Tian Y, Xu F. NK cell immunometabolism as target for liver cancer therapy. Int Immunopharmacol. 2022 Nov;112:109193.10.1016/j.intimp.2022.109193Search in Google Scholar PubMed

[21] Dodard G, Tata A, Erick TK, Jaime D, Miah SMS, Quatrini L, et al. Inflammation-induced lactate leads to rapid loss of hepatic tissue-resident NK cells. Cell Rep. 2020 Jul 7;32(1):107855.10.1016/j.celrep.2020.107855Search in Google Scholar PubMed PubMed Central

[22] Jurisić V. Characteristics of natural killer cell. Srp Arh Celok Lek. 2006;134:71–6.Search in Google Scholar

[23] Sheppard S, Santosa EK, Lau CM, Violante S, Giovanelli P, Kim H, et al. Lactate dehydrogenase A-dependent aerobic glycolysis promotes natural killer cell anti-viral and anti-tumor function. Cell Rep. 2021 Jun;35(9):109210.10.1016/j.celrep.2021.109210Search in Google Scholar PubMed PubMed Central

[24] Husain Z, Huang Y, Seth P, Sukhatme VP. Tumor-derived lactate modifies antitumor immune response: Effect on myeloid-derived suppressor cells and NK cells. J Immunol. 2013 Aug;191(3):1486–95.10.4049/jimmunol.1202702Search in Google Scholar PubMed

[25] Long Y, Gao Z, Hu X, Xiang F, Wu Z, Zhang J, et al. Downregulation of MCT4 for lactate exchange promotes the cytotoxicity of NK cells in breast carcinoma. Cancer Med. 2018 Sep;7(9):4690–700.10.1002/cam4.1713Search in Google Scholar PubMed PubMed Central

[26] Pötzl J, Roser D, Bankel L, Hömberg N, Geishauser A, Brenner CD, et al. Reversal of tumor acidosis by systemic buffering reactivates NK cells to express IFN-γ and induces NK cell-dependent lymphoma control without other immunotherapies. Int J Cancer. 2017 May;140(9):2125–33.10.1002/ijc.30646Search in Google Scholar PubMed

[27] Vaupel P, Multhoff G. Revisiting the Warburg effect: Historical dogma versus current understanding. J Physiol. 2021 Mar;599(6):1745–57.10.1113/JP278810Search in Google Scholar PubMed

[28] Murphy DA, Cheng H, Yang T, Yan X, Adjei IM. Reversing hypoxia with PLGA-encapsulated manganese dioxide nanoparticles improves natural killer cell response to tumor spheroids. Mol Pharm. 2021 Aug;18(8):2935–46.10.1021/acs.molpharmaceut.1c00085Search in Google Scholar PubMed

[29] Kumar A, Pyaram K, Yarosz EL, Hong H, Lyssiotis CA, Giri S, et al. Enhanced oxidative phosphorylation in NKT cells is essential for their survival and function. Proc Natl Acad Sci U S A. 2019 Apr;116(15):7439–48.10.1073/pnas.1901376116Search in Google Scholar PubMed PubMed Central

[30] Eshghjoo S, Kim DM, Jayaraman A, Sun Y, Alaniz RC. Macrophage polarization in atherosclerosis. Genes (Basel). 2022 Apr;13(5):756.10.3390/genes13050756Search in Google Scholar PubMed PubMed Central

[31] Roiniotis J, Dinh H, Masendycz P, Turner A, Elsegood CL, Scholz GM, et al. Hypoxia prolongs monocyte/macrophage survival and enhanced glycolysis is associated with their maturation under aerobic conditions. J Immunol. 2009 Jun;182(12):7974–81.10.4049/jimmunol.0804216Search in Google Scholar PubMed

[32] Seth P, Csizmadia E, Hedblom A, Vuerich M, Xie H, Li M, et al. Deletion of lactate dehydrogenase-A in myeloid cells triggers antitumor immunity. Cancer Res. 2017 Jul;77(13):3632–43.10.1158/0008-5472.CAN-16-2938Search in Google Scholar PubMed PubMed Central

[33] Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014 Sep;513(7519):559–63.10.1038/nature13490Search in Google Scholar PubMed PubMed Central

[34] Bohn T, Rapp S, Luther N, Klein M, Bruehl TJ, Kojima N, et al. Tumor immunoevasion via acidosis-dependent induction of regulatory tumor-associated macrophages. Nat Immunol. 2018 Dec;19(12):1319–29.10.1038/s41590-018-0226-8Search in Google Scholar PubMed

[35] Samuvel DJ, Sundararaj KP, Nareika A, Lopes-Virella MF, Huang Y. Lactate boosts TLR4 signaling and NF-kappaB pathway-mediated gene transcription in macrophages via monocarboxylate transporters and MD-2 up-regulation. J Immunol. 2009 Feb;182(4):2476–84.10.4049/jimmunol.0802059Search in Google Scholar PubMed PubMed Central

[36] Hoque R, Farooq A, Ghani A, Gorelick F, Mehal WZ. Lactate reduces liver and pancreatic injury in Toll-like receptor- and inflammasome-mediated inflammation via GPR81-mediated suppression of innate immunity. Gastroenterology. 2014 Jun;146(7):1763–74.10.1053/j.gastro.2014.03.014Search in Google Scholar PubMed PubMed Central

[37] Adam C, Paolini L, Gueguen N, Mabilleau G, Preisser L, Blanchard S, et al. Acetoacetate protects macrophages from lactic acidosis-induced mitochondrial dysfunction by metabolic reprograming. Nat Commun. 2021 Dec;12(1):7115.10.1038/s41467-021-27426-xSearch in Google Scholar PubMed PubMed Central

[38] Chen H, Yang T, Zhu L, Zhao Y. Cellular metabolism on T-cell development and function. Int Rev Immunol. 2015 Jan;34(1):19–33.10.3109/08830185.2014.902452Search in Google Scholar PubMed

[39] Comito G, Iscaro A, Bacci M, Morandi A, Ippolito L, Parri M, et al. Lactate modulates CD4+ T-cell polarization and induces an immunosuppressive environment, which sustains prostate carcinoma progression via TLR8/miR21 axis. Oncogene. 2019 May;38(19):3681–95.10.1038/s41388-019-0688-7Search in Google Scholar PubMed

[40] Peng M, Yin N, Chhangawala S, Xu K, Leslie CS, Li MO. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science. 2016 Oct;354(6311):481–4.10.1126/science.aaf6284Search in Google Scholar PubMed PubMed Central

[41] Pucino V, Certo M, Bulusu V, Cucchi D, Goldmann K, Pontarini E, et al. Lactate buildup at the site of chronic inflammation promotes disease by inducing CD4+ T cell metabolic rewiring. Cell Metab. 2019 Dec;30(6):1055–74.e8.10.1016/j.cmet.2019.10.004Search in Google Scholar PubMed PubMed Central

[42] Caslin HL, Abebayehu D, Pinette JA, Ryan JJ. Lactate is a metabolic mediator that shapes immune cell fate and function. Front Physiol. 2021 Oct;12:688485.10.3389/fphys.2021.688485Search in Google Scholar PubMed PubMed Central

[43] Haas R, Smith J, Rocher-Ros V, Nadkarni S, Montero-Melendez T, D'Acquisto F, et al. Lactate regulates metabolic and pro-inflammatory circuits in control of T cell migration and effector functions. PLoS Biol. 2015 Jul;13(7):e1002202.10.1371/journal.pbio.1002202Search in Google Scholar PubMed PubMed Central

[44] Watson MJ, Vignali PDA, Mullett SJ, Overacre-Delgoffe AE, Peralta RM, Grebinoski S, et al. Metabolic support of tumour-infiltrating regulatory T cells by lactic acid. Nature. 2021 Mar;591(7851):645–51.10.1038/s41586-020-03045-2Search in Google Scholar PubMed PubMed Central

[45] Angelin A, Gil-de-Gómez L, Dahiya S, Jiao J, Guo L, Levine MH, et al. Foxp3 Reprograms T cell metabolism to function in low-glucose, high-lactate environments. Cell Metab. 2017 Jun;25(6):1282–93.e7.10.1016/j.cmet.2016.12.018Search in Google Scholar PubMed PubMed Central

[46] Kumagai S, Koyama S, Itahashi K, Tanegashima T, Lin YT, Togashi Y, et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell. 2022 Feb;40(2):201–18.e9.10.1016/j.ccell.2022.01.001Search in Google Scholar PubMed

[47] Hermans D, Gautam S, García-Cañaveras JC, Gromer D, Mitra S, Spolski R, et al. Lactate dehydrogenase inhibition synergizes with IL-21 to promote CD8+ T cell stemness and antitumor immunity. Proc Natl Acad Sci U S A. 2020 Mar;117(11):6047–55.10.1073/pnas.1920413117Search in Google Scholar PubMed PubMed Central

[48] Xu K, Yin N, Peng M, Stamatiades EG, Shyu A, Li P, et al. Glycolysis fuels phosphoinositide 3-kinase signaling to bolster T cell immunity. Science. 2021 Jan;371(6527):405–10.10.1126/science.abb2683Search in Google Scholar PubMed PubMed Central

[49] Ippolito L, Morandi A, Giannoni E, Chiarugi P. Lactate: A metabolic driver in the tumour landscape. Trends Biochem Sci. 2019 Feb;44(2):153–66.10.1016/j.tibs.2018.10.011Search in Google Scholar PubMed

[50] Mendler AN, Hu B, Prinz PU, Kreutz M, Gottfried E, Noessner E. Tumor lactic acidosis suppresses CTL function by inhibition of p38 and JNK/c-Jun activation. Int J Cancer. 2012 Aug;131(3):633–40.10.1002/ijc.26410Search in Google Scholar PubMed

[51] Bernal W, Donaldson N, Wyncoll D, Wendon J. Blood lactate as an early predictor of outcome in paracetamol-induced acute liver failure: a cohort study. Lancet. 2002 Feb;359(9306):558–63.10.1016/S0140-6736(02)07743-7Search in Google Scholar PubMed

[52] Niederwieser T, Braunwarth E, Dasari BVM, Pufal K, Szatmary P, Hackl H, et al. Early postoperative arterial lactate concentrations to stratify risk of post-hepatectomy liver failure. Br J Surg. 2021 Nov;108(11):1360–70.10.1093/bjs/znab338Search in Google Scholar PubMed

[53] Agrawal T, Maiwall R, Rajan V, Bajpai M, Jagdish RK, Sarin SK, et al. Higher circulating natural killer cells and lower lactate levels at admission predict spontaneous survival in non-acetaminophen induced acute liver failure. Clin Immunol. 2021 Oct;231:108829. 10.1016/j.clim.2021.108829. Epub 2021 Aug 20. PMID: 34419620.Search in Google Scholar PubMed

[54] Vazquez JH, Kennon-McGill S, Byrum SD, Mackintosh SG, Jaeschke H, Williams DK, et al. Acute Liver Failure Study Group. Proteomics indicates lactate dehydrogenase is prognostic in acetaminophen-induced acute liver failure patients and reveals altered signaling pathways. Toxicol Sci. 2022 Apr;187(1):25–34.10.1093/toxsci/kfac015Search in Google Scholar PubMed PubMed Central

[55] Chen W, You J, Chen J, Zhu Y. Combining the serum lactic acid level and the lactate clearance rate into the CLIF-SOFA score for evaluating the short-term prognosis of HBV-related ACLF patients. Expert Rev Gastroenterol Hepatol. 2020 Jun;14(6):483–9.10.1080/17474124.2020.1763172Search in Google Scholar PubMed

[56] Qian J, Liu G, Wang R, Liu J, Liu Y, Liang S, et al. [Risk factors for sepsis in patients with hepatic failure]. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2019 Jul;31(7):900–2. Chinese.Search in Google Scholar

[57] Ahmed HH, De Bels D, Attou R, Honore PM, Redant S. Elevated lactic acid during ketoacidosis: Pathophysiology and management. J Transl Int Med. 2019 Oct;7(3):115–7.10.2478/jtim-2019-0024Search in Google Scholar PubMed PubMed Central

[58] Wang YH, Zhu X, Feng DY, Wu BQ. [Artificial liver support system for acute-on-chronic liver failure combined with successful liver transplantation in stage III - IV hepatic encephalopathy: An analysis of 14 cases]. Zhonghua Gan Zang Bing Za Zhi. 2018 Sep;26(9):676–9. Chinese.Search in Google Scholar

[59] Bjerring PN, Hauerberg J, Frederiksen HJ, Jorgensen L, Hansen BA, Tofteng F, et al. Cerebral glutamine concentration and lactate-pyruvate ratio in patients with acute liver failure. Neurocrit Care. 2008;9(1):3–7.10.1007/s12028-008-9060-4Search in Google Scholar PubMed

[60] Chavarria L, Romero-Giménez J, Monteagudo E, Lope-Piedrafita S, Cordoba J. Real-time assessment of ¹³C metabolism reveals an early lactate increase in the brain of rats with acute liver failure. NMR Biomed. 2015 Jan;28(1):17–23.10.1002/nbm.3226Search in Google Scholar PubMed

[61] Chartier ME, Hart L, Paganelli M, Ahmed N, Bilodeau M, Alvarez F. Successful liver transplants for liver failure associated with pyruvate kinase deficiency. Pediatrics. 2018 Apr;141(Suppl 5):S385–9.10.1542/peds.2016-3896Search in Google Scholar PubMed

[62] Ferriero R, Nusco E, De Cegli R, Carissimo A, Manco G, Brunetti-Pierri N. Pyruvate dehydrogenase complex and lactate dehydrogenase are targets for therapy of acute liver failure. J Hepatol. 2018 Aug;69(2):325–35.10.1016/j.jhep.2018.03.016Search in Google Scholar PubMed PubMed Central

[63] Wang LK, Wang LW, Li X, Han XQ, Gong ZJ. Ethyl pyruvate prevents inflammatory factors release and decreases intestinal permeability in rats with D-galactosamine-induced acute liver failure. Hepatobiliary Pancreat Dis Int. 2013 Apr;12(2):180–8.10.1016/S1499-3872(13)60029-6Search in Google Scholar

[64] Zhou Z, Wang B, Pan X, Lv J, Lou Z, Han Y, et al. Microbial metabolites indole derivatives sensitize mice to D-GalN/LPS induced-acute liver failure via the Tlr2/NF-κB pathway. Front Microbiol. 2023 Jan;13:1103998.10.3389/fmicb.2022.1103998Search in Google Scholar PubMed PubMed Central

[65] Gan X, Zhang R, Gu J, Ju Z, Wu X, Wang Q, et al. Acidic microenvironment regulates the severity of hepatic ischemia/reperfusion injury by modulating the generation and function of tregs via the PI3K-mTOR pathway. Front Immunol. 2020 Jan;10:2945.10.3389/fimmu.2019.02945Search in Google Scholar PubMed PubMed Central

[66] Su X, Gao Y, Yang R. Gut microbiota-derived tryptophan metabolites maintain gut and systemic homeostasis. Cells. 2022 Jul;11(15):2296.10.3390/cells11152296Search in Google Scholar PubMed PubMed Central

[67] Song W, Li D, Tao L, Luo Q, Chen L. Solute carrier transporters: The metabolic gatekeepers of immune cells. Acta Pharm Sin B. 2020 Jan;10(1):61–78.10.1016/j.apsb.2019.12.006Search in Google Scholar PubMed PubMed Central

[68] Ramanaviciene A, Acaite J, Ramanavicius A. Chronic caffeine intake affects lysozyme activity and immune cells in mice. J Pharm Pharmacol. 2004 May;56(5):671–6.10.1211/0022357023268Search in Google Scholar PubMed

Received: 2023-03-16
Revised: 2023-05-07
Accepted: 2023-05-17
Published Online: 2023-09-11

© 2023 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 1.5.2024 from https://www.degruyter.com/document/doi/10.1515/biol-2022-0636/html
Scroll to top button