Histone lactylation bridges metabolic reprogramming and epigenetic rewiring in driving carcinogenesis: Oncometabolite fuels oncogenic transcription

Yu Zhang , Hang Song , Meili Li , Peirong Lu

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1614

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1614 DOI: 10.1002/ctm2.1614
REVIEW

Histone lactylation bridges metabolic reprogramming and epigenetic rewiring in driving carcinogenesis: Oncometabolite fuels oncogenic transcription

Author information +
History +
PDF

Abstract

Heightened lactate production in cancer cells has been linked to various cellular mechanisms such as angiogenesis, hypoxia, macrophage polarisation and T-cell dysfunction. The lactate-induced lactylation of histone lysine residues is noteworthy, as it functions as an epigenetic modification that directly augments gene transcription from chromatin. This epigenetic modification originating from lactate effectively fosters a reliance on transcription, thereby expediting tumour progression and development. Herein, this review explores the correlation between histone lactylation and cancer characteristics, revealing histone lactylation as an innovative epigenetic process that enhances the vulnerability of cells to malignancy. Moreover, it is imperative to acknowledge the paramount importance of acknowledging innovative therapeutic methodologies for proficiently managing cancer by precisely targeting lactate signalling. This comprehensive review illuminates a crucial yet inadequately investigated aspect of histone lactylation, providing valuable insights into its clinical ramifications and prospective therapeutic interventions centred on lactylation.

Keywords

cancer / histone lactylation / metabolic reprogramming

Cite this article

Download citation ▾
Yu Zhang, Hang Song, Meili Li, Peirong Lu. Histone lactylation bridges metabolic reprogramming and epigenetic rewiring in driving carcinogenesis: Oncometabolite fuels oncogenic transcription. Clinical and Translational Medicine, 2024, 14(3): e1614 DOI:10.1002/ctm2.1614

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Ward PS, Thompson CB. Metabolic reprogramming: a cancer hallmark even Warburg did not anticipate. Cancer Cell. 2012;21(3):297-308.

[2]

Alwahsh M, Knitsch R, Marchan R, et al. Metabolic profiling of thymic epithelial tumors hints to a strong Warburg effect, glutaminolysis and precarious redox homeostasis as potential therapeutic targets. Cancers (Basel). 2022;14(6):1564.

[3]

Daverio Z, Balcerczyk A, Rautureau GJP, Panthu B. How Warburg-associated lactic acidosis rewires cancer cell energy metabolism to resist glucose deprivation. Cancers (Basel). 2023;15(5):1417.

[4]

Li W, Zhou C, Yu L, et al. Tumor-derived lactate promotes resistance to bevacizumab treatment by facilitating autophagy enhancer protein RUBCNL expression through histone H3 lysine 18 lactylation (H3K18la) in colorectal cancer. Autophagy. 2023;20(1):114-130.

[5]

Kitamura F, Semba T, Yasuda-Yoshihara N, et al. Cancer-associated fibroblasts reuse cancer-derived lactate to maintain a fibrotic and immunosuppressive microenvironment in pancreatic cancer. JCI Insight. 2023;8(20):e163022.

[6]

Dou X, Fu Q, Long Q, et al. PDK4-dependent hypercatabolism and lactate production of senescent cells promotes cancer malignancy. Nat Metab. 2023;5(11):1887-1910.

[7]

Chai P, Jia R, Li Y, et al. Regulation of epigenetic homeostasis in uveal melanoma and retinoblastoma. Prog Retin Eye Res. 2022;89:101030.

[8]

Chai P, Yu J, Wang X, Ge S, Jia R. BMP9 promotes cutaneous wound healing by activating Smad1/5 signaling pathways and cytoskeleton remodeling. Clin Transl Med. 2021;11(1):e271.

[9]

Chai P, Lan P, Li S, et al. Mechanistic insight into allosteric activation of human pyruvate carboxylase by acetyl-CoA. Mol Cell. 2022;82(21):4116-4130.e4116.

[10]

Wang T, Ye Z, Li Z, et al. Lactate-induced protein lactylation: a bridge between epigenetics and metabolic reprogramming in cancer. Cell Prolif. 2023;56(10):e13478.

[11]

Zhou J, Xu W, Wu Y, et al. GPR37 promotes colorectal cancer liver metastases by enhancing the glycolysis and histone lactylation via Hippo pathway. Oncogene. 2023;42(45):3319-3330.

[12]

Guo W, Zhou B, Bie F, et al. Single-cell RNA sequencing analysis reveals transcriptional heterogeneity of multiple primary lung cancer. Clin Transl Med. 2023;13(10):e1453.

[13]

Wong MCS, Huang J, Liang PS. Is the practice of colorectal cancer screening questionable after the NordICC trial was published? Clin Transl Med. 2023;13(10):e1365.

[14]

Zhu ZM, Huo FC, Zhang J, Shan HJ, Pei DS. Crosstalk between m6A modification and alternative splicing during cancer progression. Clin Transl Med. 2023;13(10):e1460.

[15]

He Y, Ji Z, Gong Y, et al. Numb/Parkin-directed mitochondrial fitness governs cancer cell fate via metabolic regulation of histone lactylation. Cell Rep. 2023;42(2):112033.

[16]

Morris O, Deng H, Tam C, Jasper H. Warburg-like metabolic reprogramming in aging intestinal stem cells contributes to tissue hyperplasia. Cell Rep. 2020;33(8):108423.

[17]

Visan I. Histone lactylation. Nat Immunol. 2019;20(12):1558.

[18]

Izzo LT, Wellen KE. Histone lactylation links metabolism and gene regulation. Nature. 2019;574(7779):492-493.

[19]

Zhang D, Tang Z, Huang H, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574(7779):575-580.

[20]

Sun L, Zhang Y, Yang B, et al. Lactylation of METTL16 promotes cuproptosis via m(6)A-modification on FDX1 mRNA in gastric cancer. Nat Commun. 2023;14(1):6523.

[21]

Huang ZW, Zhang XN, Zhang L, et al. STAT5 promotes PD-L1 expression by facilitating histone lactylation to drive immunosuppression in acute myeloid leukemia. Signal Transduct Target Ther. 2023;8(1):391.

[22]

Pandkar MR, Sinha S, Samaiya A, Shukla S. Oncometabolite lactate enhances breast cancer progression by orchestrating histone lactylation-dependent c-Myc expression. Transl Oncol. 2023;37:101758.

[23]

Bhat PJ, Darunte L, Kareenhalli V, Dandekar J, Kumar A. Can metabolic plasticity be a cause for cancer? Warburg–Waddington legacy revisited. Clin Epigenetics. 2011;2(2):113-122.

[24]

Siska PJ, Singer K, Evert K, Renner K, Kreutz M. The immunological Warburg effect: can a metabolic-tumor-stroma score (MeTS) guide cancer immunotherapy? Immunol Rev. 2020;295(1):187-202.

[25]

Lee CH, Gundem G, Lee W, et al. Persistent severe hyperlactatemia and metabolic derangement in lethal SDHB-mutated metastatic kidney cancer: clinical challenges and examples of extreme Warburg effect. JCO Precis Oncol. 2017:1:PO.16.00007.

[26]

Rothman DL, Shulman RG. Two transition states of the glycogen shunt and two steady states of gene expression support metabolic flexibility and the Warburg effect in cancer. Neoplasia. 2021;23(9):879-886.

[27]

Sanchez-Cenizo L, Formentini L, Aldea M, et al. Up-regulation of the ATPase inhibitory factor 1 (IF1) of the mitochondrial H+-ATP synthase in human tumors mediates the metabolic shift of cancer cells to a Warburg phenotype. J Biol Chem. 2010;285(33):25308-25313.

[28]

Mazzio EA, Boukli N, Rivera N, Soliman KF. Pericellular pH homeostasis is a primary function of the Warburg effect: inversion of metabolic systems to control lactate steady state in tumor cells. Cancer Sci. 2012;103(3):422-432.

[29]

Jiang SH, Li J, Dong FY, et al. Increased serotonin signaling contributes to the Warburg effect in pancreatic tumor cells under metabolic stress and promotes growth of pancreatic tumors in mice. Gastroenterology. 2017;153(1):277-291.e219.

[30]

Wilde L, Roche M, Domingo-Vidal M, et al. Metabolic coupling and the reverse Warburg effect in cancer: implications for novel biomarker and anticancer agent development. Semin Oncol. 2017;44(3):198-203.

[31]

Lu S, Han L, Hu X, et al. N6-methyladenosine reader IMP2 stabilizes the ZFAS1/OLA1 axis and activates the Warburg effect: implication in colorectal cancer. J Hematol Oncol. 2021;14(1):188.

[32]

Cucielo MS, Cesario RC, Silveira HS, et al. Melatonin reverses the Warburg-type metabolism and reduces mitochondrial membrane potential of ovarian cancer cells independent of MT1 receptor activation. Molecules. 2022;27(14):4350.

[33]

Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029-1033.

[34]

Jenniskens JCA, Offermans K, Simons C, et al. Energy balance-related factors and risk of colorectal cancer expressing different levels of proteins involved in the Warburg effect. Cancer Epidemiol Biomarkers Prev. 2022;31(3):633-646.

[35]

Liu S, Sun Z, Liang M, et al. An unrevealed molecular function of corannulene Buckybowl glycoconjugates in selective tumor annihilation by targeting the cancer-specific Warburg effect. Adv Sci (Weinh). 2022;9(10):e2105315.

[36]

Pitroda SP, Wakim BT, Sood RF, et al. STAT1-dependent expression of energy metabolic pathways links tumour growth and radioresistance to the Warburg effect. BMC Med. 2009;7:68.

[37]

Asgari Y, Zabihinpour Z, Salehzadeh-Yazdi A, Schreiber F, Masoudi-Nejad A. Alterations in cancer cell metabolism: the Warburg effect and metabolic adaptation. Genomics. 2015;105(5-6):275-281.

[38]

Vadivalagan C, Krishnan A, Chen SJ, et al. The Warburg effect in osteoporosis: cellular signaling and epigenetic regulation of energy metabolic events to targeting the osteocalcin for phenotypic alteration. Cell Signal. 2022;100:110488.

[39]

Chen F, Liao J, Wu P, et al. Oridonin inhibits the occurrence and development of colorectal cancer by reversing the Warburg effect via reducing PKM2 dimer formation and preventing its entry into the nucleus. Eur J Pharmacol. 2023;954:175856.

[40]

Zhong X, He X, Wang Y, et al. Warburg effect in colorectal cancer: the emerging roles in tumor microenvironment and therapeutic implications. J Hematol Oncol. 2022;15(1):160.

[41]

Lyu Y, Zhang Y, Wang Y, et al. HIF-1alpha regulated WTAP overexpression promoting the Warburg effect of ovarian cancer by m6A-dependent manner. J Immunol Res. 2022;2022:6130806.

[42]

Gerlinger M, Santos CR, Spencer-Dene B, et al. Genome-wide RNA interference analysis of renal carcinoma survival regulators identifies MCT4 as a Warburg effect metabolic target. J Pathol. 2012;227(2):146-156.

[43]

Wen H, Ting JP, O'Neill LA. A role for the NLRP3 inflammasome in metabolic diseases–did Warburg miss inflammation? Nat Immunol. 2012;13(4):352-357.

[44]

Lin L, Huang H, Liao W, et al. MACC1 supports human gastric cancer growth under metabolic stress by enhancing the Warburg effect. Oncogene. 2015;34(21):2700-2710.

[45]

Hardonniere K, Saunier E, Lemarie A, et al. The environmental carcinogen benzo[a]pyrene induces a Warburg-like metabolic reprogramming dependent on NHE1 and associated with cell survival. Sci Rep. 2016;6:30776.

[46]

Pavlides S, Tsirigos A, Vera I, et al. Transcriptional evidence for the “Reverse Warburg Effect” in human breast cancer tumor stroma and metastasis: similarities with oxidative stress, inflammation, Alzheimer's disease, and “neuron-glia metabolic coupling”. Aging (Albany NY). 2010;2(4):185-199.

[47]

Li Y, Ma H. circRNA PLOD2 promotes tumorigenesis and Warburg effect in colon cancer by the miR-513a-5p/SIX1/LDHA axis. Cell Cycle. 2022;21(23):2484-2498.

[48]

Wu H, Liang W, Han M, et al. Mechanisms regulating wound healing: functional changes in biology mediated by lactate and histone lactylation. J Cell Physiol. 2023;238(10):2243-2252.

[49]

Huang Q, Li S, Chen X, et al. Association between serum lactate dehydrogenase and lymph node metastasis in cervical cancer. Oncol Lett. 2023;26(5):482.

[50]

Chai P, Ni H, Zhang H, Fan X. The evolving functions of autophagy in ocular health: a double-edged sword. Int J Biol Sci. 2016;12(11):1332-1340.

[51]

Guido C, Whitaker-Menezes D, Capparelli C, et al. Metabolic reprogramming of cancer-associated fibroblasts by TGF-beta drives tumor growth: connecting TGF-beta signaling with “Warburg-like” cancer metabolism and L-lactate production. Cell Cycle. 2012;11(16):3019-3035.

[52]

Pan L, Feng F, Wu J, et al. Demethylzeylasteral targets lactate by inhibiting histone lactylation to suppress the tumorigenicity of liver cancer stem cells. Pharmacol Res. 2022;181:106270.

[53]

Cao Z, Xu D, Harding J, et al. Lactate oxidase nanocapsules boost T cell immunity and efficacy of cancer immunotherapy. Sci Transl Med. 2023;15(717):eadd2712.

[54]

Rastogi S, Mishra SS, Arora MK, et al. Lactate acidosis and simultaneous recruitment of TGF-beta leads to alter plasticity of hypoxic cancer cells in tumor microenvironment. Pharmacol Ther. 2023;250:108519.

[55]

Peng J, Li W, Tan N, Lai X, Jiang W, Chen G. USP47 stabilizes BACH1 to promote the Warburg effect and non-small cell lung cancer development via stimulating Hk2 and Gapdh transcription. Am J Cancer Res. 2022;12(1):91-107.

[56]

Chen IT, Aoki T, Huang YT, et al. White spot syndrome virus induces metabolic changes resembling the Warburg effect in shrimp hemocytes in the early stage of infection. J Virol. 2011;85(24):12919-12928.

[57]

Sgarbi G, Righetti R, Del Dotto V, et al. The pro-oncogenic protein IF(1) does not contribute to the Warburg effect and is not regulated by PKA in cancer cells. Biochim Biophys Acta Mol Basis Dis. 2024;1870(1):166879.

[58]

Jaworska M, Szczudlo J, Pietrzyk A, et al. The Warburg effect: a score for many instruments in the concert of cancer and cancer niche cells. Pharmacol Rep. 2023;75(4):876-890.

[59]

Frisardi V, Canovi S, Vaccaro S, Frazzi R. The significance of microenvironmental and circulating lactate in breast cancer. Int J Mol Sci. 2023;24(20):15369.

[60]

Scott DA, Richardson AD, Filipp FV, et al. Comparative metabolic flux profiling of melanoma cell lines: beyond the Warburg effect. J Biol Chem. 2011;286(49):42626-42634.

[61]

Xie F, Zhang H, Zhu K, et al. PRMT5 promotes ovarian cancer growth through enhancing Warburg effect by methylating ENO1. MedComm (2020). 2023;4(2):e245.

[62]

Kodama M, Nakayama KI. A second Warburg-like effect in cancer metabolism: the metabolic shift of glutamine-derived nitrogen: a shift in glutamine-derived nitrogen metabolism from glutaminolysis to de novo nucleotide biosynthesis contributes to malignant evolution of cancer. Bioessays. 2020;42(12):e2000169.

[63]

Galicia-Vazquez G, Aloyz R. Metabolic rewiring beyond Warburg in chronic lymphocytic leukemia: how much do we actually know? Crit Rev Oncol Hematol. 2019;134:65-70.

[64]

Shlomi T, Benyamini T, Gottlieb E, Sharan R, Ruppin E. Genome-scale metabolic modeling elucidates the role of proliferative adaptation in causing the Warburg effect. PLoS Comput Biol. 2011;7(3):e1002018.

[65]

Dai W, Shen J, Yan J, et al. Glutamine synthetase limits beta-catenin-mutated liver cancer growth by maintaining nitrogen homeostasis and suppressing mTORC1. J Clin Invest. 2022;132(24):e161408.

[66]

Encarnacion-Rosado J, Sohn ASW, Biancur DE, et al. Targeting pancreatic cancer metabolic dependencies through glutamine antagonism. Nat Cancer. 2023;5:85-99.

[67]

Quek LE, van Geldermalsen M, Guan YF, et al. Glutamine addiction promotes glucose oxidation in triple-negative breast cancer. Oncogene. 2022;41(34):4066-4078.

[68]

Linder SJ, Bernasocchi T, Martinez-Pastor B, et al. Inhibition of the proline metabolism rate-limiting enzyme P5CS allows proliferation of glutamine-restricted cancer cells. Nat Metab. 2023;5(12):2131-2147.

[69]

Blaszczak W, Williams H, Swietach P. Autoregulation of H(+)/lactate efflux prevents monocarboxylate transport (MCT) inhibitors from reducing glycolytic lactic acid production. Br J Cancer. 2022;127(7):1365-1377.

[70]

Cortes-Campos C, Elizondo R, Llanos P, Uranga RM, Nualart F, Garcia MA. MCT expression and lactate influx/efflux in tanycytes involved in glia–neuron metabolic interaction. PLoS One. 2011;6(1):e16411.

[71]

Broer S, Rahman B, Pellegri G, et al. Comparison of lactate transport in astroglial cells and monocarboxylate transporter 1 (MCT 1) expressing Xenopus laevis oocytes. Expression of two different monocarboxylate transporters in astroglial cells and neurons. J Biol Chem. 1997;272(48):30096-30102.

[72]

Lopez E, Karattil R, Nannini F, et al. Inhibition of lactate transport by MCT-1 blockade improves chimeric antigen receptor T-cell therapy against B-cell malignancies. J Immunother Cancer. 2023;11(6):e006287.

[73]

Offermans K, Jenniskens JC, Simons CC, et al. Expression of proteins associated with the Warburg-effect and survival in colorectal cancer. J Pathol Clin Res. 2022;8(2):169-180.

[74]

Gallagher FA, Woitek R, McLean MA, et al. Imaging breast cancer using hyperpolarized carbon-13 MRI. Proc Natl Acad Sci U S A. 2020;117(4):2092-2098.

[75]

Martins SF, Amorim R, Viana-Pereira M, et al. Significance of glycolytic metabolism-related protein expression in colorectal cancer, lymph node and hepatic metastasis. BMC Cancer. 2016;16:535.

[76]

Zhao H, Yan C, Hu Y, et al. Differentiated cancer cell-originated lactate promotes the self-renewal of cancer stem cells in patient-derived colorectal cancer organoids. Cancer Lett. 2020;493:236-244.

[77]

Luz MC, Perez MM, Azzalis LA, et al. Evaluation of MCT1, MCT4 and CD147 genes in peripheral blood cells of breast cancer patients and their potential use as diagnostic and prognostic markers. Int J Mol Sci. 2017;18(4):170.

[78]

Wang N, Jiang X, Zhang S, et al. Structural basis of human monocarboxylate transporter 1 inhibition by anti-cancer drug candidates. Cell. 2021;184(2):370-383.e313.

[79]

Williams JA. Using PDX for preclinical cancer drug discovery: the evolving field. J Clin Med. 2018;7(3):41.

[80]

Krajnak S, Battista MJ, Hasenburg A, Schmidt M. Metronomic chemotherapy for metastatic breast cancer. Oncol Res Treat. 2022;45(1-2):12-17.

[81]

Palakurthi B, Fross SR, Guldner IH, et al. Targeting CXCL16 and STAT1 augments immune checkpoint blockade therapy in triple-negative breast cancer. Nat Commun. 2023;14(1):2109.

[82]

Pertega-Gomes N, Felisbino S, Massie CE, et al. A glycolytic phenotype is associated with prostate cancer progression and aggressiveness: a role for monocarboxylate transporters as metabolic targets for therapy. J Pathol. 2015;236(4):517-530.

[83]

Soni VK, Shukla D, Kumar A, Vishvakarma NK. Curcumin circumvent lactate-induced chemoresistance in hepatic cancer cells through modulation of hydroxycarboxylic acid receptor-1. Int J Biochem Cell Biol. 2020;123:105752.

[84]

Brown TP, Bhattacharjee P, Ramachandran S, et al. The lactate receptor GPR81 promotes breast cancer growth via a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment. Oncogene. 2020;39(16):3292-3304.

[85]

Feng J, Yang H, Zhang Y, et al. Tumor cell-derived lactate induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer cells. Oncogene. 2017;36(42):5829-5839.

[86]

Lundo K, Trauelsen M, Pedersen SF, Schwartz TW. Why Warburg works: lactate controls immune evasion through GPR81. Cell Metab. 2020;31(4):666-668.

[87]

Cheng WY, Huynh H, Chen P, Pena-Llopis S, Wan Y. Macrophage PPARgamma inhibits Gpr132 to mediate the anti-tumor effects of rosiglitazone. Elife. 2016;5:e18501.

[88]

Nii T, Prabhu VV, Ruvolo V, et al. Imipridone ONC212 activates orphan G protein-coupled receptor GPR132 and integrated stress response in acute myeloid leukemia. Leukemia. 2019;33(12):2805-2816.

[89]

Chen P, Zuo H, Xiong H, et al. Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proc Natl Acad Sci U S A. 2017;114(3):580-585.

[90]

Lahvic JL, Ammerman M, Li P, et al. Specific oxylipins enhance vertebrate hematopoiesis via the receptor GPR132. Proc Natl Acad Sci U S A. 2018;115(37):9252-9257.

[91]

Vadevoo SMP, Gunassekaran GR, Lee C, et al. The macrophage odorant receptor Olfr78 mediates the lactate-induced M2 phenotype of tumor-associated macrophages. Proc Natl Acad Sci U S A. 2021;118(37):e2102434118.

[92]

Galle E, Wong CW, Ghosh A, et al. H3K18 lactylation marks tissue-specific active enhancers. Genome Biol. 2022;23(1):207.

[93]

Wang X, Fan W, Li N, et al. YY1 lactylation in microglia promotes angiogenesis through transcription activation-mediated upregulation of FGF2. Genome Biol. 2023;24(1):87.

[94]

Yu J, Chai P, Xie M, et al. Histone lactylation drives oncogenesis by facilitating m(6)A reader protein YTHDF2 expression in ocular melanoma. Genome Biol. 2021;22(1):85.

[95]

Yang Z, Yan C, Ma J, et al. Lactylome analysis suggests lactylation-dependent mechanisms of metabolic adaptation in hepatocellular carcinoma. Nat Metab. 2023;5(1):61-79.

[96]

Chen J, Zhang M, Liu Y, et al. Histone lactylation driven by mROS-mediated glycolytic shift promotes hypoxic pulmonary hypertension. J Mol Cell Biol. 2023;14(12):mjac073.

[97]

Wang JL, Dou XD, Cheng J, et al. Functional screening and rational design of compounds targeting GPR132 to treat diabetes. Nat Metab. 2023;5(10):1726-1746.

[98]

Jenniskens JCA, Offermans K, Simons C, et al. Energy balance-related factors in childhood and adolescence and risk of colorectal cancer expressing different levels of proteins involved in the Warburg-effect. Int J Cancer. 2022;150(11):1812-1824.

[99]

Al-Azzam N. Sirtuin 6 and metabolic genes interplay in Warburg effect in cancers. J Clin Biochem Nutr. 2020;66(3):169-175.

[100]

Pan RY, He L, Zhang J, et al. Positive feedback regulation of microglial glucose metabolism by histone H4 lysine 12 lactylation in Alzheimer's disease. Cell Metab. 2022;34(4):634-648 e636.

[101]

Zdralevic M, Vucetic M, Daher B, Marchiq I, Parks SK, Pouyssegur J. Disrupting the ‘Warburg effect’ re-routes cancer cells to OXPHOS offering a vulnerability point via ‘ferroptosis’-induced cell death. Adv Biol Regul. 2018;68:55-63.

[102]

Sun S, Xu X, Liang L, et al. Lactic acid-producing probiotic Saccharomyces cerevisiae attenuates ulcerative colitis via suppressing macrophage pyroptosis and modulating gut microbiota. Front Immunol. 2021;12:777665.

[103]

Wu J, Hu M, Jiang H, et al. Endothelial cell-derived lactate triggers bone mesenchymal stem cell histone lactylation to attenuate osteoporosis. Adv Sci (Weinh). 2023;10(31):e2301300.

[104]

Susser LI, Nguyen MA, Geoffrion M, et al. Mitochondrial fragmentation promotes inflammation resolution responses in macrophages via histone lactylation. Mol Cell Biol. 2023;43(10):531-546.

[105]

Wang N, Wang W, Wang X, et al. Histone lactylation boosts reparative gene activation post-myocardial infarction. Circ Res. 2022;131(11):893-908.

[106]

Du S, Zhang X, Jia Y, et al. Hepatocyte HSPA12A inhibits macrophage chemotaxis and activation to attenuate liver ischemia/reperfusion injury via suppressing glycolysis-mediated HMGB1 lactylation and secretion of hepatocytes. Theranostics. 2023;13(11):3856-3871.

[107]

Traxler L, Herdy JR, Stefanoni D, et al. Warburg-like metabolic transformation underlies neuronal degeneration in sporadic Alzheimer's disease. Cell Metab. 2022;34(9):1248-1263 e1246.

[108]

Flores A, Schell J, Krall AS, et al. Lactate dehydrogenase activity drives hair follicle stem cell activation. Nat Cell Biol. 2017;19(9):1017-1026.

[109]

Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019;19(8):439-453.

[110]

Li X, Chen Y, Wang T, et al. GPR81-mediated reprogramming of glucose metabolism contributes to the immune landscape in breast cancer. Discov Oncol. 2023;14(1):140.

[111]

Ma R, Li X, Gong S, et al. Dual roles of lactate in EGFR-TKI-resistant lung cancer by targeting GPR81 and MCT1. J Oncol. 2022;2022:3425841.

[112]

Menendez JA, Joven J, Cufi S, et al. The Warburg effect version 2.0: metabolic reprogramming of cancer stem cells. Cell Cycle. 2013;12(8):1166-1179.

[113]

Miao Z, Zhao X, Liu X. Hypoxia induced beta-catenin lactylation promotes the cell proliferation and stemness of colorectal cancer through the wnt signaling pathway. Exp Cell Res. 2023;422(1):113439.

[114]

Li L, Chen K, Wang T, et al. Glis1 facilitates induction of pluripotency via an epigenome–metabolome–epigenome signalling cascade. Nat Metab. 2020;2(9):882-892.

[115]

Hermans D, Gautam S, Garcia-Canaveras JC, 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;117(11):6047-6055.

[116]

Xing BC, Wang C, Ji FJ, Zhang XB. Synergistically suppressive effects on colorectal cancer cells by combination of mTOR inhibitor and glycolysis inhibitor, Oxamate. Int J Clin Exp Pathol. 2018;11(9):4439-4445.

[117]

Yu H, Yin Y, Yi Y, et al. Targeting lactate dehydrogenase A (LDHA) exerts antileukemic effects on T-cell acute lymphoblastic leukemia. Cancer Commun (Lond). 2020;40(10):501-517.

[118]

Lin J, Liu G, Chen L, Kwok HF, Lin Y. Targeting lactate-related cell cycle activities for cancer therapy. Semin Cancer Biol. 2022;86(pt 3):1231-1243.

[119]

Kim EY, Chung TW, Han CW, et al. A novel lactate dehydrogenase inhibitor, 1-(phenylseleno)-4-(trifluoromethyl) benzene, suppresses tumor growth through apoptotic cell death. Sci Rep. 2019;9(1):3969.

[120]

Friberg A, Rehwinkel H, Nguyen D, et al. Structural evidence for isoform-selective allosteric inhibition of lactate dehydrogenase A. ACS Omega. 2020;5(22):13034-13041.

[121]

Daneshmandi S, Wegiel B, Seth P. Blockade of lactate dehydrogenase-A (LDH-A) improves efficacy of anti-programmed cell death-1 (PD-1) therapy in melanoma. Cancers (Basel). 2019;11(4):450.

[122]

Jiang X, Chen Z, Zhu J, et al. E2F1 promotes Warburg effect and cancer progression via upregulating ENO2 expression in Ewing sarcoma. Mol Med Rep. 2022;26(1):237.

[123]

Pereira-Vieira J, Azevedo-Silva J, Preto A, Casal M, Queiros O. MCT1, MCT4 and CD147 expression and 3-bromopyruvate toxicity in colorectal cancer cells are modulated by the extracellular conditions. Biol Chem. 2019;400(6):787-799.

[124]

Singh M, Afonso J, Sharma D, et al. Targeting monocarboxylate transporters (MCTs) in cancer: how close are we to the clinics? Semin Cancer Biol. 2023;90:1-14.

[125]

Sprowl-Tanio S, Habowski AN, Pate KT, et al. Lactate/pyruvate transporter MCT-1 is a direct Wnt target that confers sensitivity to 3-bromopyruvate in colon cancer. Cancer Metab. 2016;4:20.

[126]

Kirkland JL, Tchkonia T. Senolytic drugs: from discovery to translation. J Intern Med. 2020;288(5):518-536.

[127]

Ghafouri-Fard S, Shabestari FA, Vaezi S, et al. Emerging impact of quercetin in the treatment of prostate cancer. Biomed Pharmacother. 2021;138:111548.

[128]

Eichner R, Heider M, Fernandez-Saiz V, et al. Immunomodulatory drugs disrupt the cereblon–CD147–MCT1 axis to exert antitumor activity and teratogenicity. Nat Med. 2016;22(7):735-743.

[129]

Afonso J, Santos LL, Miranda-Goncalves V, et al. CD147 and MCT1-potential partners in bladder cancer aggressiveness and cisplatin resistance. Mol Carcinog. 2015;54(11):1451-1466.

[130]

Xiong J, He J, Zhu J, et al. Lactylation-driven METTL3-mediated RNA m(6)A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell. 2022;82(9):1660-1677.e1610.

[131]

Zhai X, Yang Y, Wan J, Zhu R, Wu Y. Inhibition of LDH-A by oxamate induces G2/M arrest, apoptosis and increases radiosensitivity in nasopharyngeal carcinoma cells. Oncol Rep. 2013;30(6):2983-2991.

[132]

Granchi C, Roy S, Giacomelli C, et al. Discovery of N-hydroxyindole-based inhibitors of human lactate dehydrogenase isoform A (LDH-A) as starvation agents against cancer cells. J Med Chem. 2011;54(6):1599-1612.

[133]

Lin X, Zhang F, Bradbury CM, et al. 2-Deoxy-D-glucose-induced cytotoxicity and radiosensitization in tumor cells is mediated via disruptions in thiol metabolism. Cancer Res. 2003;63(12):3413-3417.

[134]

Michel KA, Zielinski R, Walker CM, et al. Hyperpolarized pyruvate MR spectroscopy depicts glycolytic inhibition in a mouse model of glioma. Radiology. 2019;293(1):168-173.

[135]

Kraus D, Reckenbeil J, Veit N, et al. Targeting glucose transport and the NAD pathway in tumor cells with STF-31: a re-evaluation. Cell Oncol (Dordr). 2018;41(5):485-494.

[136]

Wu Q, Ba-Alawi W, Deblois G, et al. GLUT1 inhibition blocks growth of RB1-positive triple negative breast cancer. Nat Commun. 2020;11(1):4205.

[137]

Clem B, Telang S, Clem A, et al. Small-molecule inhibition of 6-phosphofructo-2-kinase activity suppresses glycolytic flux and tumor growth. Mol Cancer Ther. 2008;7(1):110-120.

[138]

Thirusangu P, Ray U, Sarkar Bhattacharya S, et al. PFKFB3 regulates cancer stemness through the hippo pathway in small cell lung carcinoma. Oncogene. 2022;41(33):4003-4017.

[139]

Kashyap A, Umar SM, Dev JRA, et al. Combination of 3PO analog PFK15 and siPFKL efficiently suppresses the migration, colony formation ability, and PFK-1 activity of triple-negative breast cancers by reducing the glycolysis. J Cell Biochem. 2023;124(9):1259-1272.

[140]

Park JH, Kundu A, Lee SH, et al. Specific pyruvate kinase M2 inhibitor, compound 3K, induces autophagic cell death through disruption of the glycolysis pathway in ovarian cancer cells. Int J Biol Sci. 2021;17(8):1895-1908.

[141]

Ni M, Zhou J, Zhu Z, et al. Shikonin and cisplatin synergistically overcome cisplatin resistance of ovarian cancer by inducing ferroptosis via upregulation of HMOX1 to promote Fe(2+) accumulation. Phytomedicine. 2023;112:154701.

[142]

Wang Y, Zhang J, Wang F, Chen W, Ma J, Wang H. Alkannin inhibits the development of ovarian cancer by affecting miR-4461. Evid Based Complement Alternat Med. 2021;2021:5083302.

[143]

Jia Z, Xie Y, Wu H, et al. Phlorizin from sweet tea inhibits the progress of esophageal cancer by antagonizing the JAK2/STAT3 signaling pathway. Oncol Rep. 2021;46(1):137.

[144]

Hung MH, Chen YL, Chen LJ, et al. Canagliflozin inhibits growth of hepatocellular carcinoma via blocking glucose-influx-induced beta-catenin activation. Cell Death Dis. 2019;10(6):420.

[145]

Okada J, Yamada E, Saito T, et al. Dapagliflozin inhibits cell adhesion to collagen I and IV and increases ectodomain proteolytic cleavage of DDR1 by increasing ADAM10 activity. Molecules. 2020;25(3):495.

[146]

Yao X, Xie R, Cao Y, et al. Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J Nanobiotechnology. 2021;19(1):311.

[147]

Amorim R, Pinheiro C, Miranda-Goncalves V, et al. Monocarboxylate transport inhibition potentiates the cytotoxic effect of 5-fluorouracil in colorectal cancer cells. Cancer Lett. 2015;365(1):68-78.

[148]

George K, Thomas NS, Malathi R. 4,4′-Diisothiocyanatostilbene-2,2′-disulfonate modulates voltage-gated K(+) current and influences cell cycle arrest in androgen sensitive and insensitive human prostate cancer cell lines. Toxicol Mech Methods. 2020;30(5):358-369.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

350

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/