Lactylation in cancer: Mechanisms in tumour biology and therapeutic potentials
Yipeng He
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorTianbao Song
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorJinzhuo Ning
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorZefeng Wang
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorZhen Yin
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorPengcheng Jiang
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorQin Yuan
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorCorresponding Author
Weimin Yu
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Correspondence
Fan Cheng and Weimin Yu, Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China.
Email: [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Fan Cheng
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Correspondence
Fan Cheng and Weimin Yu, Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China.
Email: [email protected]; [email protected]
Search for more papers by this authorYipeng He
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorTianbao Song
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorJinzhuo Ning
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorZefeng Wang
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorZhen Yin
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorPengcheng Jiang
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorQin Yuan
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Search for more papers by this authorCorresponding Author
Weimin Yu
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Correspondence
Fan Cheng and Weimin Yu, Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China.
Email: [email protected]; [email protected]
Search for more papers by this authorCorresponding Author
Fan Cheng
Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
Correspondence
Fan Cheng and Weimin Yu, Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China.
Email: [email protected]; [email protected]
Search for more papers by this authorYipeng He, Tianbao Song and Jinzhuo Ning contributed equally to this study
Abstract
Lactylation, a recently identified form of protein post-translational modification (PTM), has emerged as a key player in cancer biology. The Warburg effect, a hallmark of tumour metabolism, underscores the significance of lactylation in cancer progression. By regulating gene transcription and protein function, lactylation facilitates metabolic reprogramming, enabling tumours to adapt to nutrient limitations and sustain rapid growth. Over the past decade, extensive research has revealed the intricate regulatory network underlying lactylation in tumours. Large-scale sequencing and machine learning have confirmed the widespread occurrence of lactylation sites across the tumour proteome. Targeting lactylation enzymes or metabolic pathways has demonstrated promising anti-tumour effects, highlighting the therapeutic potential of this modification. This review comprehensively explores the mechanisms of lactylation in cancer cells and the tumour microenvironment. We expound on the application of advanced omics technologies for target identification and data modelling within the lactylation field. Additionally, we summarise existing anti-lactylation drugs and discuss their clinical implications. By providing a comprehensive overview of recent advancements, this review aims to stimulate innovative research and accelerate the translation of lactylation-based therapies into clinical practice.
Key points
-
Lactylation significantly influences tumour metabolism and gene regulation, contributing to cancer progression.
-
Advanced sequencing and machine learning reveal widespread lactylation sites in tumours.
-
Targeting lactylation enzymes shows promise in enhancing anti-tumour drug efficacy and overcoming chemotherapy resistance.
-
This review outlines the clinical implications and future research directions of lactylation in oncology.
CONFLICT OF INTEREST STATEMENT
The authors declare no competing interests.
Open Research
DATA AVAILABILITY STATEMENT
No datasets were generated or analysed during the current study.
REFERENCES
- 1Maekawa M, Inomata M, Sasaki MS, et al. Electrophoretic variant of a lactate dehydrogenase isoenzyme and selective promoter methylation of the LDHA gene in a human retinoblastoma cell line. Clin Chem. 2002; 48: 1938-1945. doi:10.1093/clinchem/48.11.1938
- 2Valvona CJ, Fillmore HL, Nunn PB, Pilkington GJ. The regulation and function of lactate dehydrogenase A: therapeutic potential in brain tumor. Brain Pathol. 2015; 26: 3-17. doi:10.1111/bpa.12299
- 3Ždralević M, Brand A, Di Ianni L, et al. Double genetic disruption of lactate dehydrogenases A and B is required to ablate the “Warburg effect” restricting tumor growth to oxidative metabolism. J Biol Chem. 2018; 293: 15947-15961. doi:10.1074/jbc.ra118.004180
- 4Robergs RA, McNulty CR, Minett GM, et al. Lactate, not lactic acid, is produced by cellular cytosolic energy catabolism. Physiology (Bethesda). 2018; 33: 10-12. doi:10.1152/physiol.00033.2017
- 5Landro JA, Brush EJ, Kozarich JW. Isomerization of (R)- and (S)- glutathiolactaldehydes by glyoxalase I: the case for dichotomous stereochemical behavior in a single active site. Biochemistry. 1992; 31: 6069-6077. doi:10.1021/bi00141a016
- 6Manosalva C, Quiroga J, Hidalgo AI, et al. Role of lactate in inflammatory processes: friend or foe. Front Immunol. 2022; 12:808799. doi:10.3389/fimmu.2021.808799
- 7Zhou H, Yan X, Yu W, et al. Lactic acid in macrophage polarization: the significant role in inflammation and cancer. Int Rev Immunol. 2021; 41: 4-18. doi:10.1080/08830185.2021.1955876
- 8Lunt SY, Vander Heiden MG. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu Rev Cell Dev Biol. 2011; 27: 441-464. doi:10.1146/annurev-cellbio-092910-154237
- 9Shen Z, Jiang L, Yuan Y, et al. Inhibition of G protein-coupled receptor 81 (GPR81) protects against ischemic brain injury. CNS Neurosci Ther. 2014; 21: 271-279. doi:10.1111/cns.12362
- 10Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022; 12: 31-46. doi:10.1158/2159-8290.cd-21-1059
- 11Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011; 144: 646-674. doi:10.1016/j.cell.2011.02.013
- 12Souto-Carneiro MM, Klika KD, Abreu MT, et al. Effect of Increased lactate dehydrogenase a activity and aerobic glycolysis on the proinflammatory profile of autoimmune CD8+ T cells in rheumatoid arthritis. Arthritis Rheumatol. 2020; 72: 2050-2064. doi:10.1002/art.41420
- 13Ahmed HH, De Bels D, Attou R, et al. Elevated lactic acid during ketoacidosis: pathophysiology and management. J Transl Int Med. 2019; 7: 115-117. doi:10.2478/jtim-2019-0024
- 14Andreucci E, Margheri F, Peppicelli S, et al. OUP accepted manuscript. Reumatologia. 2021. doi:10.1093/rheumatology/keab022
10.1093/rheumatology/keab022 Google Scholar
- 15Kobayashi M, Narumi K, Furugen A, Iseki K. Transport function, regulation, and biology of human monocarboxylate transporter 1 (hMCT1) and 4 (hMCT4). Pharmacol Ther. 2021; 226:107862. doi:10.1016/j.pharmthera.2021.107862
- 16Zhao P, Wang S, Jiang J, et al. Targeting lactate metabolism and immune interaction in breast tumor via protease-triggered delivery. J Control Release. 2023; 358: 706-717. doi:10.1016/j.jconrel.2023.05.024
- 17Blaszczak 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. doi:10.1038/s41416-022-01910-7
- 18Lopez 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:e006287. doi:10.1136/jitc-2022-006287
- 19Fan M, Yang K, Wang X, et al. Lactate promotes endothelial-to-mesenchymal transition via Snail1 lactylation after myocardial infarction. Sci Adv. 2023; 9:eadc9465. doi:10.1126/sciadv.adc9465
- 20Mattei AL, Bailly N, Meissner A. DNA methylation: a historical perspective. Trends Genet. 2022; 38: 676-707. doi:10.1016/j.tig.2022.03.010
- 21Shvedunova M, Akhtar A. Modulation of cellular processes by histone and non-histone protein acetylation. Nat Rev Mol Cell Biol. 2022; 23: 329-349. doi:10.1038/s41580-021-00441-y
- 22Bilbrough T, Piemontese E, Seitz O. Dissecting the role of protein phosphorylation: a chemical biology toolbox. Chem Soc Rev. 2022; 51: 5691-5730. doi:10.1039/d1cs00991e
- 23Cockram PE, Kist M, Prakash S, et al. Ubiquitination in the regulation of inflammatory cell death and cancer. Cell Death Differ. 2021; 28: 591-605. doi:10.1038/s41418-020-00708-5
- 24Magalhães A, Duarte HO, Reis CA. The role of O-glycosylation in human disease. Mol Asp Medicine. 2021; 79:100964. doi:10.1016/j.mam.2021.100964
- 25Sabari BR, Zhang D, Allis CD, Zhao Y. Metabolic regulation of gene expression through histone acylations. Nat Rev Mol Cell Biol. 2016; 18: 90-101. doi:10.1038/nrm.2016.140
- 26Zhang D, Tang Z, Huang H, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019; 574: 575-580. doi:10.1038/s41586-019-1678-1
- 27Gaffney DO, Jennings EQ, Anderson CC, et al. Non-enzymatic lysine lactoylation of glycolytic enzymes. Cell Chem Biol. 2020; 27: 206-213. doi:10.1016/j.chembiol.2019.11.005. e6.
- 28Egeblad M, Nakasone ES, Werb Z. Tumors as organs: complex tissues that interface with the entire organism. Dev Cell. 2010; 18: 884-901. doi:10.1016/j.devcel.2010.05.012
- 29Baghban R, Roshangar L, Jahanban-Esfahlan R, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 2020; 18: 59. doi:10.1186/s12964-020-0530-4
- 30Binnewies M, Roberts EW, Kersten K, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018; 24: 541-550. doi:10.1038/s41591-018-0014-x
- 31Hu T, Cheng B, Matsunaga A, et al. Single-cell analysis defines highly specific leukemia-induced neutrophils and links MMP8 expression to recruitment of tumor associated neutrophils during FGFR1 driven leukemogenesis. Exp Hematol Oncol. 2024; 13(1): 49. doi:10.1186/s40164-024-00514-6
- 32Warburg O, Wind F, Negelein E. The metabolism OF tumors in the body. J Gen Physiol. 1927; 8: 519-530. doi:10.1085/jgp.8.6.519
- 33Brizel DM, Schroeder T, Scher RL, et al. Elevated tumor lactate concentrations predict for an increased risk of metastases in head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2001; 51: 349-353. doi:10.1016/s0360-3016(01)01630-3
- 34Brooks GA. The science and translation of lactate shuttle theory. Cell Metab. 2018; 27: 757-785. doi:10.1016/j.cmet.2018.03.008
- 35Narita T, Weinert BT, Choudhary C. Functions and mechanisms of non-histone protein acetylation. Nat Rev Mol Cell Biol. 2018; 20: 156-174. doi:10.1038/s41580-018-0081-3
- 36Barba I, Carrillo-Bosch L, Seoane J. Targeting the Warburg effect in cancer: where do we stand? IJMS. 2024; 25: 3142. doi:10.3390/ijms25063142
- 37Ding R, Yu X, Hu Z, et al. Lactate modulates RNA splicing to promote CTLA-4 expression in tumor-infiltrating regulatory T cells. Immunity. 2024; 57: 528-540. doi:10.1016/j.immuni.2024.01.019. e6.
- 38Fang X, Zhao P, Gao S, et al. Lactate induces tumor-associated macrophage polarization independent of mitochondrial pyruvate carrier-mediated metabolism. Int J Biol Macromol. 2023; 237:123810. doi:10.1016/j.ijbiomac.2023.123810
- 39Raychaudhuri D, Bhattacharya R, Sinha BP, et al. Lactate induces pro-tumor reprogramming in intratumoral plasmacytoid dendritic cells. Front Immunol. 2019; 10. doi:10.3389/fimmu.2019.01878
10.3389/fimmu.2019.01878 Google Scholar
- 40Mao Y, Zhang J, Zhou Q, et al. Hypoxia induces mitochondrial protein lactylation to limit oxidative phosphorylation. Cell Res. 2024; 34: 13-30. doi:10.1038/s41422-023-00864-6
- 41Ju J, Zhang H, Lin M, et al. The alanyl-tRNA synthetase AARS1 moonlights as a lactyl-transferase to promote YAP signaling in gastric cancer. J Clin Invest. 2024; 134(10):e174587. doi:10.1172/jci174587
- 42Zong Z, Xie F, Wang S, et al. Alanyl-tRNA synthetase, AARS1, is a lactate sensor and lactyltransferase that lactylates p53 and contributes to tumorigenesis. Cell. 2024; 187(10): 2375-2392. doi:10.1016/j.cell.2024.04.002
- 43Yang K, Fan M, Wang X, et al. Lactate promotes macrophage HMGB1 lactylation, acetylation, and exosomal release in polymicrobial sepsis. Cell Death Differ. 2021; 29(1): 133-146. doi:10.1038/s41418-021-00841-9
- 44Zessin M, Meleshin M, Praetorius L, et al. Uncovering robust delactoylase and depyruvoylase activities of HDAC isoforms. ACS Chem Biol. 2022; 17: 1364-1375. doi:10.1021/acschembio.1c00863
- 45Zu H, Li C, Dai C, et al. SIRT2 functions as a histone delactylase and inhibits the proliferation and migration of neuroblastoma cells. Cell Discov. 2022; 8: 54. doi:10.1038/s41421-022-00398-y
- 46Moreno-Yruela C, Zhang D, Wei W, et al. 2022) Class I histone deacetylases (HDAC1–3) are histone lysine delactylases. Sci Adv; 8:eabi6696. doi:10.1126/sciadv.abi6696
- 47Xie B, Zhang M, Li J, et al. KAT8-catalyzed lactylation promotes eEF1A2-mediated protein synthesis and colorectal carcinogenesis. Proc Natl Acad Sci USA. 2024; 121. doi:10.1073/pnas.2314128121
10.1073/pnas.2314128121 Google Scholar
- 48Niu Z, Chen C, Wang S, et al. HBO1 catalyzes lysine lactylation and mediates histone H3K9la to regulate gene transcription. Nat Commun. 2024; 15. doi:10.1038/s41467-024-47900-6
10.1038/s41467-024-47900-6 Google Scholar
- 49Varner EL, Trefely S, Bartee D, et al. Quantification of lactoyl-CoA (lactyl-CoA) by liquid chromatography mass spectrometry in mammalian cells and tissues. Open Biol. 2020; 10:200187. doi:10.1098/rsob.200187
- 50Zhang X-W, Li L, Liao M, et al. Thermal proteome profiling strategy identifies CNPY3 as a cellular target of gambogic acid for inducing prostate cancer pyroptosis. J Med Chem. 2024. doi:10.1021/acs.jmedchem.4c00140
10.1021/acs.jmedchem.4c00140 Google Scholar
- 51Sgarra R, Battista S, Cerchia L, et al. Mechanism of action of lactic acid on histones in cancer. Antioxid Redox Signal. 2023; 40(4-6): 236-249. doi:10.1089/ars.2022.0190
- 52Peng X, Yu Z, Surineni G, et al. Discovery of novel benzohydroxamate-based histone deacetylase 6 (HDAC6) inhibitors with the ability to potentiate anti-PD-L1 immunotherapy in melanoma. J Enzym Inhib Medicinal Chem. 2023; 38:2201408. doi:10.1080/14756366.2023.2201408
- 53Lu X, Yan G, Dawood M, et al. A novel moniliformin derivative as pan-inhibitor of histone deacetylases triggering apoptosis of leukemia cells. Biochem Pharmacol. 2021; 194:114677. doi:10.1016/j.bcp.2021.114677
- 54van den Bosch T, Boichenko A, Leus NGJ, et al. The histone acetyltransferase p300 inhibitor C646 reduces pro-inflammatory gene expression and inhibits histone deacetylases. Biochem Pharmacol. 2016; 102: 130-140. doi:10.1016/j.bcp.2015.12.010
- 55Bhattacharya A, Chatterjee S, Bhaduri U, et al. Butyrylation meets adipogenesis-probed by a p300-catalyzed acylation-specific small molecule inhibitor: implication in anti-obesity therapy. J Med Chem. 2022; 65: 12273-12291. doi:10.1021/acs.jmedchem.2c00943
- 56Li F, Si W, Xia L, et al. Positive feedback regulation between glycolysis and histone lactylation drives oncogenesis in pancreatic ductal adenocarcinoma. Mol Cancer. 2024; 23: 90. doi:10.1186/s12943-024-02008-9
- 57Xie B, Lin J, Chen X, et al. CircXRN2 suppresses tumor progression driven by histone lactylation through activating the Hippo pathway in human bladder cancer. Mol Cancer. 2023; 22: 151. doi:10.1186/s12943-023-01856-1
- 58He 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; 42:112033. doi:10.1016/j.celrep.2023.112033
- 59Guertin DA, Wellen KE. Acetyl-CoA metabolism in cancer. Nat Rev Cancer. 2023; 23: 156-172. doi:10.1038/s41568-022-00543-5
- 60Rho H, Terry AR, Chronis C, Hay N. Hexokinase 2-mediated gene expression via histone lactylation is required for hepatic stellate cell activation and liver fibrosis. Cell Metab. 2023; 35(8): 1406-1423.e8. doi:10.1016/j.cmet.2023.06.013
- 61Henry RA, Kuo Y-M, Andrews AJ. Differences in specificity and selectivity between CBP and p300 acetylation of histone H3 and H3/H4. Biochemistry. 2013; 52: 5746-5759. doi:10.1021/bi400684q
- 62Garnock-Jones KP. Panobinostat: first global approval. Drugs. 2015; 75: 695-704. doi:10.1007/s40265-015-0388-8
- 63Iyer SP, Foss FF. Romidepsin for the treatment of peripheral T-cell lymphoma. The Oncol. 2015; 20: 1084-1091. doi:10.1634/theoncologist.2015-0043
- 64Pandkar 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. doi:10.1016/j.tranon.2023.101758
- 65Meng Q, Sun H, Zhang Y, et al. Lactylation stabilizes DCBLD1 activating the pentose phosphate pathway to promote cervical cancer progression. J Exp Clin Cancer Res. 2024; 43(1): 36. doi:10.1186/s13046-024-02943-x
- 66Meng Q, Zhang Y, Sun H, et al. Human papillomavirus-16 E6 activates the pentose phosphate pathway to promote cervical cancer cell proliferation by inhibiting G6PD lactylation. Redox Biol. 2024; 71:103108. doi:10.1016/j.redox.2024.103108
- 67Yang L, Niu K, Wang J, et al. Nucleolin lactylation contributes to intrahepatic cholangiocarcinoma pathogenesis via RNA splicing regulation of MADD. J Hepatol. 2024; 81(4):651-666. doi:10.1016/j.jhep.2024.04.010
10.1016/j.jhep.2024.04.010 Google Scholar
- 68Wang S-S, Gu Y-F, Wolff N, et al. Bap1is essential for kidney function and cooperates withVhlin renal tumorigenesis. Proc Natl Acad Sci USA. 2014; 111: 16538-16543. doi:10.1073/pnas.1414789111
- 69Harlander S, Schönenberger D, Toussaint NC, et al. Combined mutation in Vhl, Trp53 and Rb1 causes clear cell renal cell carcinoma in mice. Nat Med. 2017; 23: 869-877. doi:10.1038/nm.4343
- 70Gossage L, Eisen T, Maher ER. VHL, the story of a tumour suppressor gene. Nat Rev Cancer. 2014; 15: 55-64. doi:10.1038/nrc3844
10.1038/nrc3844 Google Scholar
- 71Sato Y, Yoshizato T, Shiraishi Y, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 2013; 45: 860-867. doi:10.1038/ng.2699
- 72Yang J, Luo L, Zhao C, et al. A positive feedback loop between inactive VHL-triggered histone lactylation and PDGFRβ signaling drives clear cell renal cell carcinoma progression. Int J Biol Sci. 2022; 18: 3470-3483. doi:10.7150/ijbs.73398
- 73Zhou 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: 3319-3330. doi:10.1038/s41388-023-02841-0
- 74Wei S, Zhang J, Zhao R, et al. Histone lactylation promotes malignant progression by facilitating USP39 expression to target PI3K/AKT/HIF-1α signal pathway in endometrial carcinoma. Cell Death Discov. 2024; 10. doi:10.1038/s41420-024-01898-4
10.1038/s41420-024-01898-4 Google Scholar
- 75Zang Y, Wang A, Zhang J, et al. Hypoxia promotes histone H3K9 lactylation to enhance LAMC2 transcription in esophageal squamous cell carcinoma. iScience. 2024; 27:110188. doi:10.1016/j.isci.2024.110188
- 76Li G, Wang D, Zhai Y, et al. Glycometabolic reprogramming-induced XRCC1 lactylation confers therapeutic resistance in ALDH1A3-overexpressing glioblastoma. Cell Metab. 2024; 36: 1696-1710. doi:10.1016/j.cmet.2024.07.011. e10.
- 77Jin J, Bai L, Wang D, et al. SIRT3-dependent delactylation of cyclin E2 prevents hepatocellular carcinoma growth. EMBO Reports. 2023; 24. doi:10.15252/embr.202256052
10.15252/embr.202256052 Google Scholar
- 78Yu J, Chai P, Xie M, et al. Histone lactylation drives oncogenesis by facilitating m6A reader protein YTHDF2 expression in ocular melanoma. Genome Biol. 2021; 22: 85. doi:10.1186/s13059-021-02308-z
- 79Gu X, Zhuang A, Yu J, et al. Histone lactylation-boosted ALKBH3 potentiates tumor progression and diminished promyelocytic leukemia protein nuclear condensates by m1A demethylation of SP100A. Nucl Acids Res. 2023; 52: 2273-2289. doi:10.1093/nar/gkad1193
10.1093/nar/gkad1193 Google Scholar
- 80Chen M, Cen K, Song Y, et al. NUSAP1-LDHA-Glycolysis-Lactate feedforward loop promotes Warburg effect and metastasis in pancreatic ductal adenocarcinoma. Cancer Lett. 2023; 567:216285. doi:10.1016/j.canlet.2023.216285
- 81Huang H, Wang S, Xia H, et al. Lactate enhances NMNAT1 lactylation to sustain nuclear NAD+ salvage pathway and promote survival of pancreatic adenocarcinoma cells under glucose-deprived conditions. Cancer Lett. 2024; 588:216806. doi:10.1016/j.canlet.2024.216806
- 82Huang Y, Luo G, Peng K, et al. Lactylation stabilizes TFEB to elevate autophagy and lysosomal activity. J Cell Biol. 2024; 223. doi:10.1083/jcb.202308099
10.1083/jcb.202308099 Google Scholar
- 83Wang L, Li S, Luo H, et al. PCSK9 promotes the progression and metastasis of colon cancer cells through regulation of EMT and PI3K/AKT signaling in tumor cells and phenotypic polarization of macrophages. J Exp Clin Cancer Res. 2022; 41(1): 303. doi:10.1186/s13046-022-02477-0
- 84Li X-M, Yang Y, Jiang F-Q, et al. Histone lactylation inhibits RARγ expression in macrophages to promote colorectal tumorigenesis through activation of TRAF6-IL-6-STAT3 signaling. Cell Rep. 2024; 43:113688. doi:10.1016/j.celrep.2024.113688
- 85Sun T, Liu B, Li Y, et al. Oxamate enhances the efficacy of CAR-T therapy against glioblastoma via suppressing ectonucleotidases and CCR8 lactylation. J Exp Clin Cancer Res. 2023; 42: 253. doi:10.1186/s13046-023-02815-w
- 86Kepp O, Bezu L, Yamazaki T, et al. ATP and cancer immunosurveillance. EMBO J. 2021; 40(13):e108130. doi:10.15252/embj.2021108130
- 87Liu Y, Geng Y-H, Yang H, et al. Extracellular ATP drives breast cancer cell migration and metastasis via S100A4 production by cancer cells and fibroblasts. Cancer Lett. 2018; 430: 1-10. doi:10.1016/j.canlet.2018.04.043
- 88Yang H, Geng Y-H, Wang P, et al. Extracellular ATP promotes breast cancer chemoresistance via HIF-1α signaling. Cell Death Dis. 2022; 13. doi:10.1038/s41419-022-04647-6
10.1038/s41419?022?04647?6 Google Scholar
- 89Chiarella AM, Ryu YK, Manji GA, Rustgi AK. Extracellular ATP and adenosine in cancer pathogenesis and treatment. Trends Cancer. 2021; 7: 731-750. doi:10.1016/j.trecan.2021.04.008
- 90Yegutkin GG, Boison D. ATP and adenosine metabolism in cancer: exploitation for therapeutic gain. Pharmacol Rev. 2022; 74: 799-824. doi:10.1124/pharmrev.121.000528
- 91De Leo A, Ugolini A, Yu X, et al. Glucose-driven histone lactylation promotes the immunosuppressive activity of monocyte-derived macrophages in glioblastoma. Immunity. 2024; 57(5): 1105-1123.e8. doi:10.1016/j.immuni.2024.04.006
- 92Scheinecker C, Göschl L, Bonelli M. Treg cells in health and autoimmune diseases: new insights from single cell analysis. J Autoimmun. 2020; 110:102376. doi:10.1016/j.jaut.2019.102376
- 93Fischer L, Herkner C, Kitte R, et al. Foxp3+ regulatory T cells in bone and hematopoietic homeostasis. Front Endocrinol. 2019; 10. doi:10.3389/fendo.2019.00578
10.3389/fendo.2019.00578 Google Scholar
- 94Saito T, Nishikawa H, Wada H, et al. Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med. 2016; 22: 679-684. doi:10.1038/nm.4086
- 95Wang Z-H, Zhang P, Peng W-B, et al. Altered phenotypic and metabolic characteristics of FOXP3+CD3+CD56+ natural killer T (NKT)-like cells in human malignant pleural effusion. Oncoimmunology. 2022; 12. doi:10.1080/2162402x.2022.2160558
10.1080/2162402x.2022.2160558 Google Scholar
- 96Gu J, Zhou J, Chen Q, et al. Tumor metabolite lactate promotes tumorigenesis by modulating MOESIN lactylation and enhancing TGF-β signaling in regulatory T cells. Cell Rep. 2022; 39:110986. doi:10.1016/j.celrep.2022.110986
- 97Xiong J, He J, Zhu J, et al. Lactylation-driven METTL3-mediated RNA m6A modification promotes immunosuppression of tumor-infiltrating myeloid cells. Mol Cell. 2022; 82: 1660-1677. doi:10.1016/j.molcel.2022.02.033
- 98Li C, Teixeira AF, Zhu H-J, ten Dijke P. Cancer associated-fibroblast-derived exosomes in cancer progression. Mol Cancer. 2021; 20. doi:10.1186/s12943-021-01463-y
10.1186/s12943-021-01463-y Google Scholar
- 99Ippolito L, Comito G, Parri M, et al. Lactate rewires lipid metabolism and sustains a metabolic–epigenetic axis in prostate cancer. Cancer Res. 2022; 82: 1267-1282. doi:10.1158/0008-5472.can-21-0914
- 100Kitamura 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:e163022. doi:10.1172/jci.insight.163022
- 101Comito G, Iscaro A, Bacci 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; 38: 3681-3695. doi:10.1038/s41388-019-0688-7
- 102Mrázek J, Mekadim C, Kučerová P, et al. Melanoma-related changes in skin microbiome. Folia Microbiol. 2018; 64: 435-442. doi:10.1007/s12223-018-00670-3
- 103Gomes S, Cavadas B, Ferreira JC, et al. Profiling of lung microbiota discloses differences in adenocarcinoma and squamous cell carcinoma. Sci Rep. 2019; 9. doi:10.1038/s41598-019-49195-w
10.1038/s41598-019-49195-w Google Scholar
- 104Udayasuryan B, Zhou Z, Ahmad RN, et al. Fusobacterium nucleatum infection modulates the transcriptome and epigenome of HCT116 colorectal cancer cells in an oxygen-dependent manner. Commun Biol. 2024; 7. doi:10.1038/s42003-024-06201-w
- 105Dejea CM, Fathi P, Craig JM, et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science. 2018; 359: 592-597. doi:10.1126/science.aah3648
- 106Fu A, Yao B, Dong T, et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell. 2022; 185(8): 1356-1372. doi:10.1016/j.cell.2022.02.027
- 107Wang J, Liu Z, Xu Y, et al. Enterobacterial LPS-inducible LINC00152 is regulated by histone lactylation and promotes cancer cells invasion and migration. Front Cell Infect Microbiol. 2022; 12:913815. doi:10.3389/fcimb.2022.913815
- 108Huang Z-W, Zhang X-N, Zhang L, et al. STAT5 promotes PD-L1 expression by facilitating histone lactylation to drive immunosuppression in acute myeloid leukemia. Sig Transduct Target Ther. 2023; 8: 391. doi:10.1038/s41392-023-01605-2
- 109Wang X, Ying T, Yuan J, et al. BRAFV600E restructures cellular lactylation to promote anaplastic thyroid cancer proliferation. Endocr Relat Cancer. 2023; 30. doi:10.1530/erc-22-0344
10.1530/ERC-22-0344 Google Scholar
- 110Sun X, He L, Liu H, et al. The diapause-like colorectal cancer cells induced by SMC4 attenuation are characterized by low proliferation and chemotherapy insensitivity. Cell Metab. 2023; 35: 1563-1579. doi:10.1016/j.cmet.2023.07.005. e8.
- 111Li 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: 1-17. doi:10.1080/15548627.2023.2249762
- 112Li L, Li Z, Meng X, et al. Histone lactylation-derived LINC01127 promotes the self-renewal of glioblastoma stem cells via the cis-regulating the MAP4K4 to activate JNK pathway. Cancer Lett. 2023; 579:216467. doi:10.1016/j.canlet.2023.216467
- 113Pan 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. doi:10.1016/j.phrs.2022.106270
- 114Yu Y, Huang X, Liang C, Zhang P. Evodiamine impairs HIF1A histone lactylation to inhibit Sema3A-mediated angiogenesis and PD-L1 by inducing ferroptosis in prostate cancer. Eur J Pharmacol. 2023; 957:176007. doi:10.1016/j.ejphar.2023.176007
- 115Hong H, Chen X, Wang H, et al. Global profiling of protein lysine lactylation and potential target modified protein analysis in hepatocellular carcinoma. Proteomics. 2023; 23:e2200432. doi:10.1002/pmic.202200432
- 116Yang D, Yin J, Shan L, et al. Identification of lysine-lactylated substrates in gastric cancer cells. iScience. 2022; 25:104630. doi:10.1016/j.isci.2022.104630
- 117Cheng Z, Huang H, Li M, Chen Y. Proteomic analysis identifies PFKP lactylation in SW480 colon cancer cells. iScience. 2024; 27:108645. doi:10.1016/j.isci.2023.108645
- 118Chao J, Chen G, Huang S, et al. High histone H3K18 lactylation level is correlated with poor prognosis in epithelial ovarian cancer. Neo. 2024; 71: 319-332. doi:10.4149/neo_2024_240127n41
- 119Zhu M, Xiao Q, Cai X, et al. Predicting lymphoma prognosis using machine learning-based genes associated with lactylation. Transl Oncol. 2024; 49:102102. doi:10.1016/j.tranon.2024.102102
- 120Duan Y, Zhan H, Wang Q, et al. Integrated lactylome characterization reveals the molecular dynamics of protein regulation in gastrointestinal cancers. Adv Sci. 2024; 11(35):e2400227. doi:10.1002/advs.202400227
10.1002/advs.202400227 Google Scholar
- 121Cheng Z, Huang H, Li M, et al. Lactylation-related gene signature effectively predicts prognosis and treatment responsiveness in hepatocellular carcinoma. Pharmaceuticals. 2023; 16: 644. doi:10.3390/ph16050644
- 122Yang H, Zou X, Yang S, et al. Identification of lactylation related model to predict prognostic, tumor infiltrating immunocytes and response of immunotherapy in gastric cancer. Front Immunol. 2023; 14. doi:10.3389/fimmu.2023.1149989
10.3389/fimmu.2023.1149989 Google Scholar
- 123Sun J, Li Y, Chen R, et al. Exploring the role of <scp>lactylation-related</scp>genes in osteosarcoma: a deep dive into prognostic significance and therapeutic potential. Environ Toxicol. 2023; 39: 1001-1017. doi:10.1002/tox.24011
- 124Jiao Y, Ji F, Hou L, et al. Lactylation-related gene signature for prognostic prediction and immune infiltration analysis in breast cancer. Heliyon. 2024; 10:e24777. doi:10.1016/j.heliyon.2024.e24777
- 125Pan J, Zhang J, Lin J, et al. Constructing lactylation-related genes prognostic model to effectively predict the disease-free survival and treatment responsiveness in prostate cancer based on machine learning. Front Genet. 2024; 15. doi:10.3389/fgene.2024.1343140
10.3389/fgene.2024.1343140 Google Scholar
- 126Cai D, Yuan X, Cai DQ, et al. Integrative analysis of lactylation-related genes and establishment of a novel prognostic signature for hepatocellular carcinoma. J Cancer Res Clin Oncol. 2023; 149(13): 11517-11530. doi:10.1007/s00432-023-04947-0
- 127Meyer KD, Saletore Y, Zumbo P, et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell. 2012; 149: 1635-1646. doi:10.1016/j.cell.2012.05.003
- 128Yang L, Wang X, Liu J, et al. Prognostic and tumor microenvironmental feature of clear cell renal cell carcinoma revealed by m6A and lactylation modification-related genes. Front Immunol. 2023; 14:1225023. doi:10.3389/fimmu.2023.1225023
- 129Zheng P, Mao Z, Luo M, et al. Comprehensive bioinformatics analysis of the solute carrier family and preliminary exploration of SLC25A29 in lung adenocarcinoma. Cancer Cell Int. 2023; 23(1): 222. doi:10.1186/s12935-023-03082-7
- 130Melliou E, Chinou I. Chemistry and bioactivity of royal jelly from Greece. J Agric Food Chem. 2005; 53: 8987-8992. doi:10.1021/jf051550p
- 131Xu H, Li L, Wang S, et al. Royal jelly acid suppresses hepatocellular carcinoma tumorigenicity by inhibiting H3 histone lactylation at H3K9la and H3K14la sites. Phytomedicine. 2023; 118:154940. doi:10.1016/j.phymed.2023.154940
- 132Geng Y, Zheng X, Zhang D, et al. CircHIF1A induces cetuximab resistance in colorectal cancer by promoting HIF1α-mediated glycometabolism alteration. Biol Direct. 2024; 19. doi:10.1186/s13062-024-00478-x
- 133Huang Q, Chu Z, Wang Z, et al. circCDK13-loaded small extracellular vesicles accelerate healing in preclinical diabetic wound models. Nat Commun. 2024; 15. doi:10.1038/s41467-024-48284-3
10.1038/s41467?024?48284?3 Google Scholar
- 134Ng AYE, Chan SN, Pek JW. Genetic compensation between ribosomal protein paralogs mediated by a cognate circular RNA. Cell Rep. 2024; 43:114228. doi:10.1016/j.celrep.2024.114228
- 135Zhang Z, Wang Y, Zhang Y, et al. The function and mechanisms of action of circular RNAs in urologic cancer. Mol Cancer. 2023; 22: 61. doi:10.1186/s12943-023-01766-2
- 136Zhou C, Li W, Liang Z, et al. Mutant KRAS-activated circATXN7 fosters tumor immunoescape by sensitizing tumor-specific T cells to activation-induced cell death. Nat Commun. 2024; 15: 499. doi:10.1038/s41467-024-44779-1
- 137Samuel M, Gabrielsson S. Personalized medicine and back–allogeneic exosomes for cancer immunotherapy. J Intern Med. 2019; 289(2): 138-146. doi:10.1111/joim.12963
- 138Morand S, Devanaboyina M, Staats H, et al. Ovarian cancer immunotherapy and personalized medicine. IJMS. 2021; 22: 6532. doi:10.3390/ijms22126532
- 139Certo M, Tsai C-H, Pucino V, et al. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol. 2020; 21: 151-161. doi:10.1038/s41577-020-0406-2
- 140Xie X-Q, Yang Y, Wang Q, et al. Targeting ATAD3A-PINK1-mitophagy axis overcomes chemoimmunotherapy resistance by redirecting PD-L1 to mitochondria. Cell Res. 2023; 33(3): 215-228. doi:10.1038/s41422-022-00766-z
- 141Basile G, Bandini M, Gibb EA, et al. Neoadjuvant pembrolizumab and radical cystectomy in patients with muscle-invasive urothelial bladder cancer: 3-year median follow-up update of PURE-01 trial. Clin Cancer Res. 2022; 28: 5107-5114. doi:10.1158/1078-0432.ccr-22-2158
- 142Peyraud F, Guégan J-P, Bodet D, et al. Circulating L-arginine predicts the survival of cancer patients treated with immune checkpoint inhibitors. Ann Oncol. 2022; 33: 1041-1051. doi:10.1016/j.annonc.2022.07.001
- 143Castro A, Pyke RM, Zhang X, et al. Strength of immune selection in tumors varies with sex and age. Nat Commun. 2020; 11. doi:10.1038/s41467-020-17981-0
10.1038/s41467-020-17981-0 Google Scholar
- 144Mehla K, Singh PK. Metabolic regulation of macrophage polarization in cancer. Trends Cancer. 2019; 5: 822-834. doi:10.1016/j.trecan.2019.10.007
- 145MANTOVANI A, SOZZANI S, LOCATI M, et al. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002; 23: 549-555. doi:10.1016/s1471-4906(02)02302-5
- 146Chaudagar K, Hieromnimon HM, Kelley A, et al. Suppression of tumor cell lactate-generating signaling pathways eradicates murine PTEN/p53-deficient aggressive-variant prostate cancer via macrophage phagocytosis. bioRxiv. 2023.
- 147Kabir AU, Subramanian M, Lee DH, et al. Dual role of endothelial Myct1 in tumor angiogenesis and tumor immunity. Sci Transl Med. 2021; 13:eabb6731. doi:10.1126/scitranslmed.abb6731
- 148Li X, Peng X, Zhang C, et al. Bladder cancer-derived small extracellular vesicles promote tumor angiogenesis by inducing hbp-related metabolic reprogramming and SerRS O-GlcNAcylation in endothelial cells. Adv Sci. 2022:2202993. doi:10.1002/advs.202202993
10.1002/advs.202202993 Google Scholar
- 149Quintanal-Villalonga A, Taniguchi H, Zhan YA, et al. Multi-omic analysis of lung tumors defines pathways activated in neuroendocrine transformation. bioRxiv. 2020.
- 150Aggarwal R, Huang J, Alumkal JJ, et al. Clinical and genomic characterization of treatment-emergent small-cell neuroendocrine prostate cancer: a multi-institutional prospective study. JCO. 2018; 36: 2492-2503. doi:10.1200/jco.2017.77.6880
- 151Marcoux N, Gettinger SN, O'Kane G, et al. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: clinical outcomes. JCO. 2019; 37: 278-285. doi:10.1200/jco.18.01585
- 152Wang D, Du G, Chen X, et al. Zeb1-controlled metabolic plasticity enables remodeling of chromatin accessibility in the development of neuroendocrine prostate cancer. Cell Death Differ. 2024; 31(6): 779-791. doi:10.1038/s41418-024-01295-5
- 153Duan W, Liu W, Xia S, et al. Warburg effect enhanced by AKR1B10 promotes acquired resistance to pemetrexed in lung cancer-derived brain metastasis. J Transl Med. 2023; 21. doi:10.1186/s12967-023-04403-0
10.1186/s12967-023-04403-0 Google Scholar
- 154Chu Y-D, Cheng L-C, Lim S-N, et al. Aldolase B-driven lactagenesis and CEACAM6 activation promote cell renewal and chemoresistance in colorectal cancer through the Warburg effect. Cell Death Dis. 2023; 14: 660. doi:10.1038/s41419-023-06187-z
- 155Hondo E, Stewart C. Profiling gene expression in growth-arrested mouse embryos in diapause. Genome Biology. 2004;6: 202. doi:10.1186/gb-2004-6-1-202
- 156Renfree MB, Shaw G. Diapause. Annu Rev Physiol. 2000; 62: 353-375. doi:10.1146/annurev.physiol.62.1.353
- 157Rehman SK, Haynes J, Collignon E, et al. Colorectal cancer cells enter a diapause-like DTP state to survive chemotherapy. Cell. 2021; 184: 226-242. doi:10.1016/j.cell.2020.11.018. e21.
- 158Dhimolea E, de Matos Simoes R, Kansara D, et al. An embryonic diapause-like adaptation with suppressed myc activity enables tumor treatment persistence. Cancer Cell. 2021; 39: 240-256. doi:10.1016/j.ccell.2020.12.002. e11.
- 159Sansbury BM, Kmiec EB. On the origins of homology directed repair in mammalian cells. IJMS. 2021; 22: 3348. doi:10.3390/ijms22073348
- 160Russo M, Crisafulli G, Sogari A, et al. Adaptive mutability of colorectal cancers in response to targeted therapies. Science. 2019; 366: 1473-1480. doi:10.1126/science.aav4474
- 161Ray Chaudhuri A, Callen E, Ding X, et al. Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature. 2016; 5: 382-387. doi:10.1038/nature18325
10.1038/nature18325 Google Scholar
- 162Chen G, Chen J, Qiao Y, et al. ZNF830 mediates cancer chemoresistance through promoting homologous-recombination repair. Nucl Acids Res. 2017; 46: 1266-1279. doi:10.1093/nar/gkx1258
10.1093/nar/gkx1258 Google Scholar
- 163Chen Y, Wu J, Zhai L, et al. Metabolic regulation of homologous recombination repair by MRE11 lactylation. Cell. 2023; 187(2): 294-311.e21. doi:10.1016/j.cell.2023.11.022
- 164Zafar A, Wang W, Liu G, et al. Molecular targeting therapies for neuroblastoma: progress and challenges. Med Res Rev. 2020; 41: 961-1021. doi:10.1002/med.21750
- 165Shariati M, Meric-Bernstam F. Targeting AKT for cancer therapy. Expert Opin Investig Drug. 2019; 28: 977-988. doi:10.1080/13543784.2019.1676726
- 166Dudley AC, Griffioen AW. Pathological angiogenesis: mechanisms and therapeutic strategies. Angiogenesis. 2023; 26: 313-347. doi:10.1007/s10456-023-09876-7
- 167Liu Y, Huang N, Liao S, et al. Current research progress in targeted anti-angiogenesis therapy for osteosarcoma. Cell Prolif. 2021; 54. doi:10.1111/cpr.13102
10.1111/cpr.13102 Google Scholar
- 168Yetkin-Arik B, Kastelein AW, Klaassen I, et al. Angiogenesis in gynecological cancers and the options for anti-angiogenesis therapy. Biochim Biophys Acta (BBA Rev Cancer. 2021; 1875:188446. doi:10.1016/j.bbcan.2020.188446
- 169Zhang L, Xu J, Zhou S, et al. Endothelial DGKG promotes tumor angiogenesis and immune evasion in hepatocellular carcinoma. J Hepatol. 2024; 80: 82-98. doi:10.1016/j.jhep.2023.10.006
- 170Garcia J, Hurwitz HI, Sandler AB, et al. Bevacizumab (Avastin®) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 2020; 86:102017. doi:10.1016/j.ctrv.2020.102017
- 171Maione F, Capano S, Regano D, et al. Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice. J Clin Invest. 2012; 122: 1832-1848. doi:10.1172/jci58976
- 172Panda M, Tripathi SK, Zengin G, Biswal BK. Evodiamine as an anticancer agent: a comprehensive review on its therapeutic application, pharmacokinetic, toxicity, and metabolism in various cancers. Cell Biol Toxicol. 2022; 39: 1-31. doi:10.1007/s10565-022-09772-8
- 173Ohanna M, Cheli Y, Bonet C, et al. Secretome from senescent melanoma engages the STAT3 pathway to favor reprogramming of naive melanoma towards a tumor-initiating cell phenotype. Oncotarget. 2013; 4: 2212-2224. doi:10.18632/oncotarget.1143
- 174Iliopoulos D, Hirsch HA, Wang G, Struhl K. Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion. Proc Natl Acad Sci USA. 2011; 108: 1397-1402. doi:10.1073/pnas.1018898108
- 175Giannoni E, Bianchini F, Masieri L, et al. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Res. 2010; 70: 6945-6956. doi:10.1158/0008-5472.can-10-0785
- 176Vermeulen L, De Sousa E Melo F, van der Heijden M, et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010; 12: 468-476. doi:10.1038/ncb2048
- 177Li M, Nishimura T, Takeuchi Y, et al. FXYD3 functionally demarcates an ancestral breast cancer stem cell subpopulation with features of drug-tolerant persisters. J Clin Invest. 2023; 133. doi:10.1172/jci166666
10.1172/JCI166666 Google Scholar
- 178Jia L, Zhang W, Wang C-Y. BMI1 inhibition eliminates residual cancer stem cells after PD1 blockade and activates antitumor immunity to prevent metastasis and relapse. Cell Stem Cell. 2020; 27: 238-253. doi:10.1016/j.stem.2020.06.022. e6.
- 179Miao Z, Zhao X, Liu X. Hypoxia induced β-catenin lactylation promotes the cell proliferation and stemness of colorectal cancer through the wnt signaling pathway. Exp Cell Res. 2023; 422:113439. doi:10.1016/j.yexcr.2022.113439