Understanding the Contribution of Lactate Metabolism in Cancer Progress: A Perspective from Isomers
Abstract
:Simple Summary
Abstract
1. Introduction
2. Lactate Metabolism in Carcinoma Cells
2.1. Warburg Effect
2.2. Metabolism of Lactate Isomers and Aerobic Glycolysis
2.2.1. L-Lactate Production in Aerobic Glycolysis
2.2.2. D-Lactate Production in Aerobic Glycolysis
3. Current Advances of Lactate in Cancer
3.1. Breast Cancer
3.2. Cervical Cancer
3.3. Lung Cancer
3.4. Pancreatic Cancer
3.5. Prostate Cancer
3.6. Liver Cancer
4. Lactate Metabolism Related Prognostic Markers in Cancer
4.1. LDH
4.2. Glyoxalases
4.3. MCTs
4.4. GPR81
4.5. GPR132
5. The Downstream Molecular Signalings of Lactate (L-and D-Lactate) Mediation in Cancer
5.1. MAPK
5.2. HIF-1α
5.3. NDRG3
5.4. PI3K/AKT
5.5. NF-κB
5.6. Wnt Signaling
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koppenol, W.H.; Bounds, P.L.; Dang, C.V. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat. Rev. Cancer 2011, 11, 325–337. [Google Scholar] [CrossRef] [PubMed]
- Hui, S.; Ghergurovich, J.M.; Morscher, R.J.; Jang, C.; Teng, X.; Lu, W.; Esparza, L.A.; Reya, T.; Zhan, L.; White, E.; et al. Glucose feeds the TCA cycle via circulating lactate. Nature 2017, 551, 115–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirschhaeuser, F.; Sattler, U.G.; Mueller-Klieser, W. Lactate: A metabolic key player in cancer. Cancer Res. 2011, 71, 6921–6925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walenta, S.; Schroeder, T.; Mueller-Klieser, W. Lactate in solid malignant tumors: Potential basis of a metabolic classification in clinical oncology. Curr. Med. Chem. 2004, 11, 2195–2204. [Google Scholar] [CrossRef]
- Gatenby, R.A.; Gillies, R.J. Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer 2004, 4, 891–899. [Google Scholar] [CrossRef]
- Walenta, S.; Mueller-Klieser, W.F. Lactate: Mirror and motor of tumor malignancy. Semin. Radiat. Oncol. 2004, 14, 267–274. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- De Berardinis, R.J.; Lum, J.J.; Hatzivassiliou, G.; Thompson, C.B. The biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008, 7, 11–20. [Google Scholar] [CrossRef] [Green Version]
- Warbug, O. The metabolism of carcinoma cells. J. Cancer Res. 1925, 9, 148–163. [Google Scholar] [CrossRef]
- Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cardenas, C.; Lovy, A.; Silva-Pavez, E.; Urra, F.; Mizzoni, C.; Ahumada-Castro, U.; Bustos, G.; Jaňa, F.; Cruz, P.; Foskett, J.K.; et al. Cancer cells with defective oxidative phosphorylation require endoplasmic reticulum-to-mitochondria Ca(2+) transfer for survival. Sci. Signal. 2020, 13, eaay1212. [Google Scholar] [CrossRef] [PubMed]
- Pavlova, N.N.; Thompson, C.B. The Emerging Hallmarks of Cancer Metabolism. Cell Metab. 2016, 23, 27–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weinberg, F.; Hamanaka, R.; Wheaton, W.W.; Weinberg, S.; Joseph, J.; Lopez, M.; Kalyanaraman, B.; Mutlu, G.M.; Budinger, G.R.S.; Chandel, N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA 2010, 107, 8788–8793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bonnay, F.; Veloso, A.; Steinmann, V.; Kocher, T.; Abdusselamoglu, M.D.; Bajaj, S.; Rivelles, E.; Landskron, L.; Esterbauer, H.; Zinzen, R.P.; et al. Oxidative Metabolism Drives Immortalization of Neural Stem Cells during Tumorigenesis. Cell 2020, 182, 1490–1507.e19. [Google Scholar] [CrossRef]
- Li, T.; Han, J.; Jia, L.; Hu, X.; Chen, L.; Wang, Y. PKM2 coordinates glycolysis with mitochondrial fusion and oxidative phosphorylation. Protein Cell 2019, 10, 583–594. [Google Scholar] [CrossRef] [Green Version]
- Birts, C.N.; Banerjee, A.; Darley, M.; Dunlop, C.R.; Nelson, S.; Nijjar, S.K.; Blaydes, J.P. p53 is regulated by aerobic glycolysis in cancer cells by the CtBP family of NADH-dependent transcriptional regulators. Sci. Signal. 2020, 13, eaau9529. [Google Scholar] [CrossRef]
- Patra, K.C.; Hay, N. The pentose phosphate pathway and cancer. Trends Biochem. Sci. 2014, 39, 347–354. [Google Scholar] [CrossRef] [Green Version]
- Yamagata, M.; Hasuda, K.; Stamato, T.; Tannock, I.F. The contribution of lactic acid to acidification of tumours: Studies of variant cells lacking lactate dehydrogenase. Br. J. Cancer 1998, 77, 1726–1731. [Google Scholar] [CrossRef] [Green Version]
- Faubert, B.; Li, K.Y.; Cai, L.; Hensley, C.T.; Kim, J.; Zacharias, L.G.; Yang, C.; Do, Q.N.; Doucette, S.; Burguete, D.; et al. Lactate Metabolism in Human Lung Tumors. Cell 2017, 171, 358–371.e9. [Google Scholar] [CrossRef]
- Wang, L.; Bi, R.; Yin, H.; Liu, H.; Li, L. ENO1 silencing impaires hypoxia-induced gemcitabine chemoresistance associated with redox modulation in pancreatic cancer cells. Am. J. Transl. Res. 2019, 11, 4470–4480. [Google Scholar] [PubMed]
- Zhang, M.; Liang, L.; He, J.; He, Z.; Yue, C.; Jin, X.; Gao, M.; Xiao, S.; Zhou, Y. Fra-1 Inhibits Cell Growth and the Warburg Effect in Cervical Cancer Cells via STAT1 Regulation of the p53 Signaling Pathway. Front. Cell Dev. Biol. 2020, 8, 579629. [Google Scholar] [CrossRef] [PubMed]
- Zhou, S.; Zheng, Q.; Huang, X.; Wang, Y.; Luo, S.; Jiang, R.; Wang, L.; Ye, W.; Tian, H. Isolation and identification of l/d-lactate-conjugated bufadienolides from toad eggs revealing lactate racemization in amphibians. Org. Biomol. Chem. 2017, 15, 5609–5615. [Google Scholar] [CrossRef] [PubMed]
- Mustiere, R.; Vanelle, P.; Primas, N. Plasmodial Kinase Inhibitors Targeting Malaria: Recent Developments. Molecules 2020, 25, 5949. [Google Scholar] [CrossRef]
- Karsten, V.; Murray, S.R.; Pike, J.; Troy, K.; Ittensohn, M.; Kondradzhyan, M.; Low, K.B.; Bermudes, D. msbB deletion confers acute sensitivity to CO2 in Salmonella enterica serovar Typhimurium that can be suppressed by a loss-of-function mutation in zwf. BMC Microbiol. 2009, 9, 170. [Google Scholar] [CrossRef] [Green Version]
- Jiang, P.; Du, W.; Wang, X.; Mancuso, A.; Gao, X.; Wu, M.; Yang, X. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat. Cell Biol. 2011, 13, 310–316. [Google Scholar] [CrossRef] [Green Version]
- Peng, M.; Li, S.; He, Q.; Zhao, J.; Li, L.; Ma, H. Proteomics reveals changes in hepatic proteins during chicken embryonic development: An alternative model to study human obesity. BMC Genom. 2018, 19, 29. [Google Scholar] [CrossRef] [Green Version]
- Tiwari, S.; Mishra, M.; Salemi, M.R.; Phinney, B.S.; Newens, J.L.; Gomes, A.V. Gender-specific changes in energy metabolism and protein degradation as major pathways affected in livers of mice treated with ibuprofen. Sci. Rep. 2020, 10, 3386. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.M.; Lin, C.E.; Chen, H.H.; Cheng, Y.F.; Cheng, H.W.; Imai, K. Effect of prednisolone on glyoxalase 1 in an inbred mouse model of aristolochic acid nephropathy using a proteomics method with fluorogenic derivatization-liquid chromatography-tandem mass spectrometry. PLoS ONE 2020, 15, e0227838. [Google Scholar] [CrossRef] [Green Version]
- Pichitpunpong, C.; Thongkorn, S.; Kanlayaprasit, S.; Yuwattana, W.; Plaingam, W.; Sangsuthum, S.; Aizat, W.M.; Baharum, S.N.; Tencomnao, T.; Hu, V.W.; et al. Phenotypic subgrouping and multi-omics analyses reveal reduced diazepam-binding inhibitor (DBI) protein levels in autism spectrum disorder with severe language impairment. PLoS ONE 2019, 14, e0214198. [Google Scholar] [CrossRef]
- Gueugneau, M.; Coudy-Gandilhon, C.; Chambon, C.; Verney, J.; Taillandier, D.; Combaret, L.; Polge, C.; Walrand, S.; Roche, F.; Barthélémy, J.-C.; et al. Muscle Proteomic and Transcriptomic Profiling of Healthy Aging and Metabolic Syndrome in Men. Int. J. Mol. Sci. 2021, 22, 4205. [Google Scholar] [CrossRef] [PubMed]
- Yukimoto, R.; Nishida, N.; Hata, T.; Fujino, S.; Ogino, T.; Miyoshi, N.; Takahashi, H.; Uemura, M.; Satoh, T.; Hirofumi, Y.; et al. Specific activation of glycolytic enzyme enolase 2 in BRAF V600E-mutated colorectal cancer. Cancer Sci. 2021, 112, 2884–2894. [Google Scholar] [CrossRef] [PubMed]
- Liu, K.; Hu, H.; Wang, W.; Zhang, X. Genetic engineering of Pseudomonas chlororaphis GP72 for the enhanced production of 2-Hydroxyphenazine. Microb. Cell Fact. 2016, 15, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, Y.E.; Jeon, H.J.; Kim, D.; Lee, S.Y.; Kim, K.Y.; Hong, J.; Maeng, P.J.; Kim, K.-R.; Kang, D. Quantitative Proteomic Analysis of 2D and 3D Cultured Colorectal Cancer Cells: Profiling of Tankyrase Inhibitor XAV939-Induced Proteome. Sci. Rep. 2018, 8, 13255. [Google Scholar] [CrossRef] [Green Version]
- Zheng, X.; Boyer, L.; Jin, M.; Mertens, J.; Kim, Y.; Ma, L.; Hamm, M.; Gage, F.H.; Hunter, T. Metabolic reprogramming during neuronal differentiation from aerobic glycolysis to neuronal oxidative phosphorylation. Elife 2016, 5, e13374. [Google Scholar] [CrossRef]
- Chiarugi, A.; Dolle, C.; Felici, R.; Ziegler, M. The NAD metabolome--a key determinant of cancer cell biology. Nat. Rev. Cancer 2012, 12, 741–752. [Google Scholar] [CrossRef]
- Velez, J.; Velasquez, Z.; Silva, L.M.R.; Gartner, U.; Failing, K.; Daugschies, A.; Mazurek, S.; Hermosilla, C.; Taubert, A. Metabolic Signatures of Cryptosporidium parvum-Infected HCT-8 Cells and Impact of Selected Metabolic Inhibitors on C. parvum Infection under Physioxia and Hyperoxia. Biology 2021, 10, 60. [Google Scholar] [CrossRef]
- Deng, Y.; Song, P.; Chen, X.; Huang, Y.; Hong, L.; Jin, Q.; Ji, J. 3-Bromopyruvate-Conjugated Nanoplatform-Induced Pro-Death Autophagy for Enhanced Photodynamic Therapy against Hypoxic Tumor. ACS Nano 2020, 14, 9711–9727. [Google Scholar] [CrossRef]
- Patgiri, A.; Skinner, O.S.; Miyazaki, Y.; Schleifer, G.; Marutani, E.; Shah, H.; Sharma, R.; Goodman, R.P.; To, T.L.; Bao, X.R.; et al. An engineered enzyme that targets circulating lactate to alleviate intracellular NADH:NAD(+) imbalance. Nat. Biotechnol. 2020, 38, 309–313. [Google Scholar] [CrossRef]
- Li, Y.L.; Zhou, B.W.; Cao, Y.Q.; Zhang, J.; Zhang, L.; Guo, Y.L. Chiral Analysis of Lactate during Direct Contact Coculture by Single-Cell On-Probe Enzymatic Dehydrogenation Derivatization: Unraveling Metabolic Changes Caused by d-Lactate. Anal. Chem. 2021, 93, 4576–4583. [Google Scholar] [CrossRef]
- Tekkök, S.B.; Brown, A.M.; Westenbroek, R.; Pellerin, L.; Ransom, B.R. Transfer of glycogen-derived lactate from astrocytes to axons via specific monocarboxylate transporters supports mouse optic nerve activity. J. Neurosci. Res. 2005, 81, 644–652. [Google Scholar] [CrossRef] [PubMed]
- Ling, B.; Peng, F.; Alcorn, J.; Lohmann, K.; Bandy, B.; Zello, G.A. D-Lactate altered mitochondrial energy production in rat brain and heart but not liver. Nutr. Metab. 2012, 9, 6. [Google Scholar] [CrossRef] [Green Version]
- Connor, H.; Woods, H.F.; Ledingham, J.G.G. Comparison of the kinetics and utilisation of D(-)-and L(+)-sodium lactate in normal man. Ann. Nutr. Metab. 1983, 27, 481–487. [Google Scholar] [CrossRef] [PubMed]
- Finsterwald, C.; Magistretti, P.J.; Lengacher, S. Astrocytes: New Targets for the Treatment of Neurodegenerative Diseases. Curr. Pharm. Des. 2015, 21, 3570–3581. [Google Scholar] [CrossRef] [PubMed]
- Thornalley, P.J. Glutathione-dependent detoxification of alpha-oxoaldehydes by the glyoxalase system: Involvement in disease mechanisms and antiproliferative activity of glyoxalase I inhibitors. Chem. Biol. Interact. 1998, 111–112, 137–151. [Google Scholar] [CrossRef]
- Adeva-Andany, M.; López-Ojén, M.; Funcasta-Calderón, R.; Ameneiros-Rodríguez, E.; Donapetry-García, C.; Vila-Altesor, M.; Rodríguez-Seijas, J. Comprehensive review on lactate metabolism in human health. Mitochondrion 2014, 17, 76–100. [Google Scholar] [CrossRef]
- Cooper, R.A.; Anderson, A. The formation and catabolism of methylglyoxal during glycolysis in Escherichia coli. FEBS Lett. 1970, 11, 273–276. [Google Scholar] [CrossRef] [Green Version]
- Santel, T.; Pflug, G.; Hemdan, N.Y.; Schafer, A.; Hollenbach, M.; Buchold, M.; Hintersdorf, A.; Lindner, I.; Otto, A.; Bigl, M.; et al. Curcumin inhibits glyoxalase 1: A possible link to its anti-inflammatory and anti-tumor activity. PLoS ONE 2008, 3, e3508. [Google Scholar] [CrossRef] [Green Version]
- Bellier, J.; Nokin, M.J.; Larde, E.; Karoyan, P.; Peulen, O.; Castronovo, V.; Bellahcène, A. Methylglyoxal, a potent inducer of AGEs, connects between diabetes and cancer. Diabetes Res. Clin. Pract. 2019, 148, 200–211. [Google Scholar] [CrossRef]
- Pun, P.B.; Murphy, M.P. Pathological significance of mitochondrial glycation. Int. J. Cell Biol. 2012, 2012, 843505. [Google Scholar] [CrossRef]
- Morcos, M.; Du, X.; Pfisterer, F.; Hutter, H.; Sayed, A.A.; Thornalley, P.; Ahmed, N.; Baynes, J.; Thorpe, S.; Kukudov, G.; et al. Glyoxalase-1 prevents mitochondrial protein modification and enhances lifespan in Caenorhabditis elegans. Aging Cell 2008, 7, 260–269. [Google Scholar] [CrossRef] [PubMed]
- Birkenmeier, G.; Stegemann, C.; Hoffmann, R.; Gunther, R.; Huse, K.; Birkemeyer, C. Posttranslational modification of human glyoxalase 1 indicates redox-dependent regulation. PLoS ONE 2010, 5, e10399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Bari, L.; Moro, L.; Passarella, S. Prostate cancer cells metabolize d-lactate inside mitochondria via a D-lactate dehydrogenase which is more active and highly expressed than in normal cells. FEBS Lett. 2013, 587, 467–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fahad Ullah, M. Breast Cancer: Current Perspectives on the Disease Status. Adv. Exp. Med. Biol. 2019, 1152, 51–64. [Google Scholar]
- Pagani, O.; Senkus, E.; Wood, W.; Colleoni, M.; Cufer, T.; Kyriakides, S.; Costa, A.; Winer, E.P. International guidelines for management of metastatic breast cancer: Can metastatic breast cancer be cured? J. Natl. Cancer Inst. 2010, 102, 456–463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Veronesi, U.; Boyle, P.; Goldhirsch, A.; Orecchia, R.; Viale, G. Breast cancer. Lancet 2005, 365, 1727–1741. [Google Scholar] [CrossRef]
- Liang, Y.; Zhang, H.; Song, X.; Yang, Q. Metastatic heterogeneity of breast cancer: Molecular mechanism and potential therapeutic targets. Semin. Cancer Biol. 2020, 60, 14–27. [Google Scholar] [CrossRef]
- Cheung, S.M.; Husain, E.; Masannat, Y.; Miller, I.D.; Wahle, K.; Heys, S.D.; He, J. Lactate concentration in breast cancer using advanced magnetic resonance spectroscopy. Br. J. Cancer 2020, 123, 261–267. [Google Scholar] [CrossRef]
- Kalezic, A.; Udicki, M.; Srdic Galic, B.; Aleksic, M.; Korac, A.; Jankovic, A.; Korac, B. Lactate Metabolism in Breast Cancer Microenvironment: Contribution Focused on Associated Adipose Tissue and Obesity. Int. J. Mol. Sci. 2020, 21, 9676. [Google Scholar] [CrossRef]
- Ghergurovich, J.M.; Lang, J.D.; Levin, M.K.; Briones, N.; Facista, S.J.; Mueller, C.; Cowan, A.J.; McBride, M.J.; Rodriguez, E.S.R.; Killian, A.; et al. Local production of lactate, ribose phosphate, and amino acids within human triple-negative breast cancer. Med 2021, 2, 736–754. [Google Scholar] [CrossRef]
- Becker, L.M.; O’Connell, J.T.; Vo, A.P.; Cain, M.P.; Tampe, D.; Bizarro, L.; Sugimoto, H.; McGow, A.K.; Asara, J.M.; Lovisa, S.; et al. Epigenetic Reprogramming of Cancer-Associated Fibroblasts Deregulates Glucose Metabolism and Facilitates Progression of Breast Cancer. Cell Rep. 2020, 31, 107701. [Google Scholar] [CrossRef] [PubMed]
- Waks, A.G.; Winer, E.P. Breast Cancer Treatment: A Review. Jama 2019, 321, 288–300. [Google Scholar] [CrossRef] [PubMed]
- Guedes, M.; Araujo, J.R.; Correia-Branco, A.; Gregorio, I.; Martel, F.; Keating, E. Modulation of the uptake of critical nutrients by breast cancer cells by lactate: Impact on cell survival, proliferation and migration. Exp. Cell Res. 2016, 341, 111–122. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.J.; Shin, K.J.; Park, S.A.; Park, K.S.; Park, S.; Heo, K.; Seo, Y.K.; Noh, D.Y.; Ryu, S.O.; Suh, P.G. G-protein-coupled receptor 81 promotes a malignant phenotype in breast cancer through angiogenic factor secretion. Oncotarget 2016, 7, 70898–70911. [Google Scholar] [CrossRef] [Green Version]
- Stäubert, C.; Broom, O.J.; Nordström, A. Hydroxycarboxylic acid receptors are essential for breast cancer cells to control their lipid/fatty acid metabolism. Oncotarget 2015, 6, 19706–19720. [Google Scholar] [CrossRef] [Green Version]
- Ishihara, S.; Hata, K.; Hirose, K.; Okui, T.; Toyosawa, S.; Uzawa, N.; Nishimura, R.; Yoneda, T. The lactate sensor GPR81 regulates glycolysis and tumor growth of breast cancer. Sci. Rep. 2022, 12, 6261. [Google Scholar] [CrossRef]
- Chen, P.; Zuo, H.; Xiong, H.; Kolar, M.J.; Chu, Q.; Saghatelian, A.; Siegwart, D.J.; Wan, Y. Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proc. Natl. Acad. Sci. USA 2017, 114, 580–585. [Google Scholar] [CrossRef] [Green Version]
- Chen, F.; Chen, J.; Yang, L.; Liu, J.; Zhang, X.; Zhang, Y.; Tu, Q.; Yin, D.; Lin, D.; Wong, P.P.; et al. Extracellular vesicle-packaged HIF-1alpha-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat. Cell Biol. 2019, 21, 498–510. [Google Scholar] [CrossRef]
- Jeong, H.; Kim, S.; Hong, B.J.; Lee, C.J.; Kim, Y.E.; Bok, S.; Oh, J.M.; Gwak, S.H.; Yoo, M.Y.; Lee, M.S.; et al. Tumor-Associated Macrophages Enhance Tumor Hypoxia and Aerobic Glycolysis. Cancer Res. 2019, 79, 795–806. [Google Scholar] [CrossRef] [Green Version]
- Cohen, P.A.; Jhingran, A.; Oaknin, A.; Denny, L. Cervical cancer. Lancet 2019, 393, 169–182. [Google Scholar] [CrossRef]
- Stone, S.C.; Rossetti, R.A.M.; Alvarez, K.L.F.; Carvalho, J.P.; Margarido, P.F.R.; Baracat, E.C.; Tacla, M.; Boccardo, E.; Yokochi, K.; Lorenzi, N.P.; et al. Lactate secreted by cervical cancer cells modulates macrophage phenotype. J. Leukoc. Biol. 2019, 105, 1041–1054. [Google Scholar] [CrossRef] [PubMed]
- Boskey, E.R.; Cone, R.A.; Whaley, K.J.; Moench, T.R. Origins of vaginal acidity: High D/L lactate ratio is consistent with bacteria being the primary source. Hum. Reprod. 2001, 16, 1809–1813. [Google Scholar] [CrossRef] [PubMed]
- Wagner, W.; Ciszewski, W.M.; Kania, K.D. L- and D-lactate enhance DNA repair and modulate the resistance of cervical carcinoma cells to anticancer drugs via histone deacetylase inhibition and hydroxycarboxylic acid receptor 1 activation. Cell Commun. Signal. CCS 2015, 13, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wagner, W.; Kania, K.D.; Blauz, A.; Ciszewski, W.M. The lactate receptor (hcar1/gpr81) contributes to doxorubicin chemoresistance via abcb1 transporter up-regulation in human cervical cancer hela cells. J. Physiol. Pharmacol. 2017, 68, 555–564. [Google Scholar] [PubMed]
- Wagner, W.; Sobierajska, K.; Kania, K.D.; Paradowska, E.; Ciszewski, W.M. Lactate Suppresses Retroviral Transduction in Cervical Epithelial Cells through DNA-PKcs Modulation. Int. J. Mol. Sci. 2021, 22, 13194. [Google Scholar] [CrossRef] [PubMed]
- Wagner, W.; Kania, K.D.; Ciszewski, W.M. Stimulation of lactate receptor (HCAR1) affects cellular DNA repair capacity. DNA Repair 2017, 52, 49–58. [Google Scholar] [CrossRef]
- Da, Q.; Yan, Z.; Li, Z.; Han, Z.; Ren, M.; Huang, L.; Zhang, X.; Liu, J.; Wang, T. TAK1 is involved in sodium L-lactate-stimulated p38 signaling and promotes apoptosis. Mol. Cell. Biochem. 2021, 476, 873–882. [Google Scholar] [CrossRef]
- Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. CA Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [Green Version]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
- Nooreldeen, R.; Bach, H. Current and Future Development in Lung Cancer Diagnosis. Int. J. Mol. Sci. 2021, 22, 8661. [Google Scholar] [CrossRef]
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Fan, T.W.; Lane, A.N.; Higashi, R.M.; Farag, M.A.; Gao, H.; Bousamra, M.; Miller, D.M. Altered regulation of metabolic pathways in human lung cancer discerned by (13)C stable isotope-resolved metabolomics (SIRM). Mol. Cancer 2009, 8, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Updegraff, B.L.; Zhou, X.; Guo, Y.; Padanad, M.S.; Chen, P.H.; Yang, C.; Sudderth, J.; Rodriguez-Tirado, C.; Girard, L.; Minna, J.D.; et al. Transmembrane Protease TMPRSS11B Promotes Lung Cancer Growth by Enhancing Lactate Export and Glycolytic Metabolism. Cell reports 2018, 25, 2223–2233.e6. [Google Scholar] [CrossRef] [Green Version]
- Dong, Q.; Zhou, C.; Ren, H.; Zhang, Z.; Cheng, F.; Xiong, Z.; Wu, Z. Lactate-induced MRP1 expression contributes to metabolism-based etoposide resistance in non-small cell lung cancer cells. Cell Commun. Signal. 2020, 18, 167. [Google Scholar] [CrossRef]
- Liao, Z.X.; Kempson, I.M.; Hsieh, C.C.; Tseng, S.J.; Yang, P.C. Potential therapeutics using tumor-secreted lactate in nonsmall cell lung cancer. Drug Discov. Today 2021, 26, 2508–2514. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Yang, H.; Zhang, Y.; Wei, H.; Zhu, Z.; Zhu, B.; Yang, M.; Cao, W.; Wang, L.; Wu, Z. Tumor cell-derived lactate induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer cells. Oncogene 2017, 36, 5829–5839. [Google Scholar] [CrossRef]
- Caronni, N.; Simoncello, F.; Stafetta, F.; Guarnaccia, C.; Ruiz-Moreno, J.S.; Opitz, B.; Galli, T.; Proux-Gillardeaux, V.; Benvenuti, F. Downregulation of Membrane Trafficking Proteins and Lactate Conditioning Determine Loss of Dendritic Cell Function in Lung Cancer. Cancer Res. 2018, 78, 1685–1699. [Google Scholar] [CrossRef] [Green Version]
- Geeraerts, X.; Fernandez-Garcia, J.; Hartmann, F.J.; de Goede, K.E.; Martens, L.; Elkrim, Y.; Debraekeleer, A.; Stijlemans, B.; Vandekeere, A.; Rinaldi, G.; et al. Macrophages are metabolically heterogeneous within the tumor microenvironment. Cell Rep. 2021, 37, 110171. [Google Scholar] [CrossRef]
- Hu, M.; Zhao, Y.; Cao, Y.; Tang, Q.; Feng, Z.; Ni, J.; Zhou, X. DRP1 promotes lactate utilization in KRAS-mutant non-small-cell lung cancer cells. Cancer Sci. 2020, 111, 3588–3599. [Google Scholar] [CrossRef]
- Tauffenberger, A.; Fiumelli, H.; Almustafa, S.; Magistretti, P.J. Lactate and pyruvate promote oxidative stress resistance through hormetic ROS signaling. Cell Death Dis. 2019, 10, 653. [Google Scholar] [CrossRef] [Green Version]
- Hashimoto, T.; Hussien, R.; Oommen, S.; Gohil, K.; Brooks, G.A. Lactate sensitive transcription factor network in L6 cells: Activation of MCT1 and mitochondrial biogenesis. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2007, 21, 2602–2612. [Google Scholar] [CrossRef] [PubMed]
- Tempero, M.A. NCCN Guidelines Updates: Pancreatic Cancer. J. Natl. Compr. Cancer Netw. 2019, 17, 603–605. [Google Scholar]
- Vincent, A.; Herman, J.; Schulick, R.; Hruban, R.H.; Goggins, M. Pancreatic cancer. Lancet 2011, 378, 607–620. [Google Scholar] [CrossRef] [PubMed]
- Guillaumond, F.; Leca, J.; Olivares, O.; Lavaut, M.-N.; Vidal, N.; Berthezène, P.; Dusetti, N.J.; Loncle, C.; Calvo, E.; Turrini, O.; et al. Strengthened glycolysis under hypoxia supports tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma. Proc. Natl. Acad. Sci. USA 2013, 110, 3919–3924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhagat, T.D.; Von Ahrens, D.; Dawlaty, M.; Zou, Y.; Baddour, J.; Achreja, A.; Verma, A. Lactate-mediated epigenetic reprogramming regulates formation of human pancreatic cancer-associated fibroblasts. eLife 2019, 8, e50663. [Google Scholar] [CrossRef]
- Kumstel, S.; Schreiber, T.; Goldstein, L.; Stenzel, J.; Lindner, T.; Joksch, M.; Zhang, X.; Wendt, E.H.U.; Schönrogge, M.; Krause, B.; et al. Targeting pancreatic cancer with combinatorial treatment of CPI-613 and inhibitors of lactate metabolism. PLoS ONE 2022, 17, e0266601. [Google Scholar] [CrossRef]
- Roland, C.L.; Arumugam, T.; Deng, D.; Liu, S.H.; Philip, B.; Gomez, S.; Burns, W.R.; Ramachandran, V.; Wang, H.; Cruz-Monserrate, Z.; et al. Cell surface lactate receptor GPR81 is crucial for cancer cell survival. Cancer Res. 2014, 74, 5301–5310. [Google Scholar] [CrossRef] [Green Version]
- Yamada, Y.; Beltran, H. The treatment landscape of metastatic prostate cancer. Cancer Lett. 2021, 519, 20–29. [Google Scholar] [CrossRef]
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [Green Version]
- Ippolito, L.; Comito, G.; Parri, M.; Iozzo, M.; Duatti, A.; Virgilio, F.; Lorito, N.; Bacci, M.; Pardella, E.; Sandrini, G.; et al. Lactate Rewires Lipid Metabolism and Sustains a Metabolic-Epigenetic Axis in Prostate Cancer. Cancer Res. 2022, 82, 1267–1282. [Google Scholar] [CrossRef]
- Pertega-Gomes, N.; Baltazar, F. Lactate transporters in the context of prostate cancer metabolism: What do we know? Int. J. Mol. Sci. 2014, 15, 18333–18348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiaschi, T.; Marini, A.; Giannoni, E.; Taddei, M.L.; Gandellini, P.; De Donatis, A.; Lanciotti, M.; Serni, S.; Cirri, P.; Chiarugi, P. Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res. 2012, 72, 5130–5140. [Google Scholar] [CrossRef] [PubMed]
- Zacharias, N.; Lee, J.; Ramachandran, S.; Shanmugavelandy, S.; McHenry, J.; Dutta, P.; Millward, S.; Gammon, S.; Efstathiou, E.; Troncoso, P.; et al. Androgen Receptor Signaling in Castration-Resistant Prostate Cancer Alters Hyperpolarized Pyruvate to Lactate Conversion and Lactate Levels In Vivo. Mol. Imag. Biol. 2019, 21, 86–94. [Google Scholar] [CrossRef] [PubMed]
- Brauer, H.A.; Makowski, L.; Hoadley, K.A.; Casbas-Hernandez, P.; Lang, L.J.; Roman-Perez, E.; D’Arcy, M.; Freemerman, A.J.; Perou, C.M.; Troester, M.A. Impact of tumor microenvironment and epithelial phenotypes on metabolism in breast cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013, 19, 571–585. [Google Scholar] [CrossRef] [Green Version]
- De Bari, L.; Chieppa, G.; Marra, E.; Passarella, S. L-lactate metabolism can occur in normal and cancer prostate cells via the novel mitochondrial L-lactate dehydrogenase. Int. J. Oncol. 2010, 37, 1607–1620. [Google Scholar]
- Flick, M.J.; Konieczny, S.F. Identification of putative mammalian D-lactate dehydrogenase enzymes. Biochem. Biophys. Res. Commun. 2002, 295, 910–916. [Google Scholar] [CrossRef]
- de Bari, L.; Atlante, A.; Guaragnella, N.; Principato, G.; Passarella, S. D-Lactate transport and metabolism in rat liver mitochondria. Biochem. J. 2002, 365, 391–403. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Wang, H. Heterogeneity of liver cancer and personalized therapy. Cancer Lett. 2016, 379, 191–197. [Google Scholar] [CrossRef]
- Marengo, A.; Rosso, C.; Bugianesi, E. Liver Cancer: Connections with Obesity, Fatty Liver, and Cirrhosis. Annu. Rev. Med. 2016, 67, 103–117. [Google Scholar] [CrossRef]
- Broadfield, L.A.; Duarte, J.A.G.; Schmieder, R.; Broekaert, D.; Veys, K.; Planque, M.; Vriens, K.; Karasawa, Y.; Napolitano, F.; Fujita, S.; et al. Fat Induces Glucose Metabolism in Nontransformed Liver Cells and Promotes Liver Tumorigenesis. Cancer Res. 2021, 81, 1988–2001. [Google Scholar] [CrossRef]
- Zhao, Y.; Li, M.; Yao, X.; Fei, Y.; Lin, Z.; Li, Z.; Cai, K.; Zhao, Y.; Luo, Z. HCAR1/MCT1 Regulates Tumor Ferroptosis through the Lactate-Mediated AMPK-SCD1 Activity and Its Therapeutic Implications. Cell Rep. 2020, 33, 108487. [Google Scholar] [CrossRef] [PubMed]
- Kumagai, S.; Koyama, S.; Itahashi, K.; Tanegashima, T.; Lin, Y.T.; Togashi, Y.; Kamada, T.; Irie, T.; Okumura, G.; Kono, H.; et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell 2022, 40, 201–218.e9. [Google Scholar] [CrossRef] [PubMed]
- Niu, Y.; Lin, Z.; Wan, A.; Sun, L.; Yan, S.; Liang, H.; Zhan, S.; Chen, D.; Bu, X.; Liu, P.; et al. Loss-of-Function Genetic Screening Identifies Aldolase A as an Essential Driver for Liver Cancer Cell Growth Under Hypoxia. Hepatology 2021, 74, 1461–1479. [Google Scholar] [CrossRef] [PubMed]
- Guo, W.; Qiu, Z.; Wang, Z.; Wang, Q.; Tan, N.; Chen, T.; Chen, Z.; Huang, S.; Gu, J.; Li, J.; et al. MiR-199a-5p is negatively associated with malignancies and regulates glycolysis and lactate production by targeting hexokinase 2 in liver cancer. Hepatology 2015, 62, 1132–1144. [Google Scholar] [CrossRef]
- Lee, D.C.; Sohn, H.A.; Park, Z.-Y.; Oh, S.; Kang, Y.K.; Lee, K.-M.; Kang, M.; Jang, Y.J.; Yang, S.-J.; Hong, Y.K.; et al. A lactate-induced response to hypoxia. Cell 2015, 161, 595–609. [Google Scholar] [CrossRef] [Green Version]
- Martel, F.; Guedes, M.; Keating, E. Effect of polyphenols on glucose and lactate transport by breast cancer cells. Breast Cancer Res. Treat. 2016, 157, 1–11. [Google Scholar] [CrossRef]
- Webb, B.A.; Chimenti, M.; Jacobson, M.P.; Barber, D.L. Dysregulated pH: A perfect storm for cancer progression. Nat. Rev. Cancer 2011, 11, 671–677. [Google Scholar] [CrossRef]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef] [Green Version]
- Burgner, J.W., 2nd; Ray, W.J., Jr. On the origin of the lactate dehydrogenase induced rate effect. Biochemistry 1984, 23, 3636–3648. [Google Scholar] [CrossRef]
- Wang, Y.; Li, G.; Wan, F.; Dai, B.; Ye, D. Prognostic value of D-lactate dehydrogenase in patients with clear cell renal cell carcinoma. Oncol. Lett. 2018, 16, 866–874. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Z.; Sheng, B.; Ma, C.; Zhang, H.; Gao, C.; Su, F.; Xu, P. Relative catalytic efficiency of ldhL- and ldhD-encoded products is crucial for optical purity of lactic acid produced by lactobacillus strains. Appl. Environ. Microbiol. 2012, 78, 3480–3483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiang, J.; Zhou, L.; Zhuang, Y.; Zhang, J.; Sun, Y.; Li, S.; Zhang, Z.; Zhang, G.; He, Y. Lactate dehydrogenase is correlated with clinical stage and grade and is downregulated by siSATauB1 in ovarian cancer. Oncol. Rep. 2018, 40, 2788–2797. [Google Scholar] [PubMed]
- Ye, Y.; Chen, M.; Chen, X.; Xiao, J.; Liao, L.; Lin, F. Clinical Significance and Prognostic Value of Lactate Dehydrogenase Expression in Cervical Cancer. Genet. Test Mol. Biomark. 2022, 26, 107–117. [Google Scholar] [CrossRef]
- Liu, L.; He, Y.; Ge, G.; Zhou, P.; Zhu, Y.; Tang, H.; Huang, Y.; Li, W.; Zhang, L. Lactate dehydrogenase and creatine kinase as poor prognostic factors in lung cancer: A retrospective observational study. PLoS ONE 2017, 12, e0182168. [Google Scholar] [CrossRef] [Green Version]
- Hsieh, A.H.; Tahkar, H.; Koczwara, B.; Kichenadasse, G.; Beckmann, K.; Karapetis, C.; Sukumaran, S. Pre-treatment serum lactate dehydrogenase as a biomarker in small cell lung cancer. Asia-Pac. J. Clin. Oncol. 2018, 14, e64–e70. [Google Scholar] [CrossRef]
- Conteduca, V.; Oromendia, C.; Eng, K.W.; Bareja, R.; Sigouros, M.; Molina, A.; Faltas, B.M.; Sboner, A.; Mosquera, J.M.; Elemento, O.; et al. Clinical features of neuroendocrine prostate cancer. Eur. J. Cancer 2019, 121, 7–18. [Google Scholar] [CrossRef]
- Facchinelli, D.; Sina, S.; Boninsegna, E.; Borin, A.; Tisi, M.C.; Piazza, F.; Scapinello, G.; Maiolo, E.; Hohaus, S.; Zamò, A.; et al. Primary pancreatic lymphoma: Clinical presentation, diagnosis, treatment, and outcome. Eur. J. Haematol. 2020, 105, 468–475. [Google Scholar] [CrossRef]
- Armstrong, A.J.; George, D.J.; Halabi, S. Serum lactate dehydrogenase predicts for overall survival benefit in patients with metastatic renal cell carcinoma treated with inhibition of mammalian target of rapamycin. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2012, 30, 3402–3407. [Google Scholar] [CrossRef] [PubMed]
- Certo, M.; Tsai, C.-H.; Pucino, V.; Ho, P.-C.; Mauro, C. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat. Rev. Immunol. 2021, 21, 151–161. [Google Scholar] [CrossRef]
- Urbanska, K.; Orzechowski, A. Unappreciated Role of LDHA and LDHB to Control Apoptosis and Autophagy in Tumor Cells. Int. J. Mol. Sci. 2019, 20, 2085. [Google Scholar] [CrossRef] [Green Version]
- Doherty, J.R.; Cleveland, J.L. Targeting lactate metabolism for cancer therapeutics. J. Clin. Investig. 2013, 123, 3685–3692. [Google Scholar] [CrossRef] [PubMed]
- Khajah, M.A.; Khushaish, S.; Luqmani, Y.A. Lactate Dehydrogenase A or B Knockdown Reduces Lactate Production and Inhibits Breast Cancer Cell Motility in vitro. Front. Pharmacol. 2021, 12, 747001. [Google Scholar] [CrossRef] [PubMed]
- Zdralevic, M.; Brand, A.; Di Ianni, L.; Dettmer, K.; Reinders, J.; Singer, K.; Peter, K.; Schnell, A.; Bruss, C.; Decking, S.M.; 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. [Google Scholar] [CrossRef] [PubMed]
- Fantin, V.R.; St-Pierre, J.; Leder, P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 2006, 9, 425–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koukourakis, M.I.; Giatromanolaki, A.; Simopoulos, C.; Polychronidis, A.; Sivridis, E. Lactate dehydrogenase 5 (LDH5) relates to up-regulated hypoxia inducible factor pathway and metastasis in colorectal cancer. Clin. Exp. Metastasis 2005, 22, 25–30. [Google Scholar] [CrossRef]
- Xu, K.; Yin, N.; Peng, M.; Stamatiades, E.G.; Shyu, A.; Li, P.; Zhang, X.; Do, M.H.; Wang, Z.; Capistrano, K.J.; et al. Glycolysis fuels phosphoinositide 3-kinase signaling to bolster T cell immunity. Science 2021, 371, 405–410. [Google Scholar] [CrossRef]
- Liu, Y.; Guo, J.Z.; Liu, Y.; Wang, K.; Ding, W.; Wang, H.; Liu, X.; Zhou, S.; Lu, S.C.; Yang, H.B.; et al. Nuclear lactate dehydrogenase A senses ROS to produce alpha-hydroxybutyrate for HPV-induced cervical tumor growth. Nat. Commun. 2018, 9, 4429. [Google Scholar] [CrossRef] [Green Version]
- Le, A.; Cooper, C.R.; Gouw, A.M.; Dinavahi, R.; Maitra, A.; Deck, L.M.; Royer, R.E.; Vander Jagt, D.L.; Semenza, G.L.; Dang, C.V. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc. Natl. Acad. Sci. USA 2010, 107, 2037–2042. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.-Y.; Loo, T.Y.; Shen, J.-G.; Wang, N.; Wang, D.-M.; Yang, D.-P.; Mo, S.-L.; Guan, X.-Y.; Chen, J.-P. LDH-A silencing suppresses breast cancer tumorigenicity through induction of oxidative stress mediated mitochondrial pathway apoptosis. Breast Cancer Res. Treat. 2012, 131, 791–800. [Google Scholar] [CrossRef]
- Xie, H.; Hanai, J.-I.; Ren, J.-G.; Kats, L.; Burgess, K.; Bhargava, P.; Signoretti, S.; Billiard, J.; Duffy, K.J.; Grant, A.; et al. Targeting lactate dehydrogenase--a inhibits tumorigenesis and tumor progression in mouse models of lung cancer and impacts tumor-initiating cells. Cell Metab. 2014, 19, 795–809. [Google Scholar] [CrossRef] [Green Version]
- Al-Salam, S.; Kandhan, K.; Sudhadevi, M. Down regulation of lactate dehydrogenase initiates apoptosis in HeLa and MCF-7 cancer cells through increased voltage-dependent anion channel protein and inhibition of BCL2. Oncotarget 2021, 12, 923–935. [Google Scholar] [CrossRef] [PubMed]
- Angelin, A.; Gil-de-Gomez, L.; Dahiya, S.; Jiao, J.; Guo, L.; Levine, M.H.; Wang, Z.; Quinn, W.J.; Kopinski, P.K.; Wang, L.; et al. Foxp3 Reprograms T Cell Metabolism to Function in Low-Glucose, High-Lactate Environments. Cell Metab. 2017, 25, 1282–1293.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Billiard, J.; Dennison, J.B.; Briand, J.; Annan, R.S.; Chai, D.; Colón, M.; Dodson, C.S.; Gilbert, S.A.; Greshock, J.; Jing, J.; et al. Quinoline 3-sulfonamides inhibit lactate dehydrogenase A and reverse aerobic glycolysis in cancer cells. Cancer Metab. 2013, 1, 19. [Google Scholar] [CrossRef] [PubMed]
- Song, Y.J.; Kim, A.; Kim, G.T.; Yu, H.Y.; Lee, E.S.; Park, M.J.; Kim, Y.J.; Shim, S.M.; Park, T.S. Inhibition of lactate dehydrogenase A suppresses inflammatory response in RAW 264.7 macrophages. Mol. Med. Rep. 2019, 19, 629–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lei, W.; Kang, W.; Nan, Y.; Lei, Z.; Li, Z.; Demin, L.; Sun, L.; Huang, H. The Downregulation of miR-200c Promotes Lactate Dehydrogenase A Expression and Non-Small Cell Lung Cancer Progression. Oncol. Res. 2018, 26, 1015–1022. [Google Scholar] [CrossRef]
- Xiao, X.; Huang, X.; Ye, F.; Chen, B.; Song, C.; Wen, J.; Zhang, Z.; Zheng, G.; Tang, H.; Xie, X. The miR-34a-LDHA axis regulates glucose metabolism and tumor growth in breast cancer. Sci. Rep. 2016, 6, 21735. [Google Scholar] [CrossRef] [Green Version]
- Luo, Y.; Yang, Z.; Li, D.; Liu, Z.; Yang, L.; Zou, Q.; Yuan, Y. LDHB and FABP4 are Associated With Progression and Poor Prognosis of Pancreatic Ductal Adenocarcinomas. Appl. Immunohistochem. Mol. Morphol. 2017, 25, 351–357. [Google Scholar] [CrossRef]
- Wu, G.; Yuan, S.; Chen, Z.; Chen, G.; Fan, Q.; Dong, H.; Ye, F.; Li, J.; Zhu, X. The KLF14 Transcription Factor Regulates Glycolysis by Downregulating LDHB in Colorectal Cancer. Int. J. Biol. Sci. 2019, 15, 628–635. [Google Scholar] [CrossRef] [Green Version]
- Shi, L.; Yan, H.; An, S.; Shen, M.; Jia, W.; Zhang, R.; Zhao, L.; Huang, G. SIRT5-mediated deacetylation of LDHB promotes autophagy and tumorigenesis in colorectal cancer. Mol. Oncol. 2019, 13, 358–375. [Google Scholar] [CrossRef]
- Cheng, A.; Zhang, P.; Wang, B.; Yang, D.; Duan, X.; Jiang, Y.; Xu, T.; Jiang, Y.; Shi, J.; Ding, C.; et al. Aurora-A mediated phosphorylation of LDHB promotes glycolysis and tumor progression by relieving the substrate-inhibition effect. Nat. Commun. 2019, 10, 5566. [Google Scholar] [CrossRef] [Green Version]
- Kumar, S.; Xie, H.; Scicluna, P.; Lee, L.; Bjornhagen, V.; Hoog, A.; Larsson, C.; Lui, W.O. MiR-375 Regulation of LDHB Plays Distinct Roles in Polyomavirus-Positive and -Negative Merkel Cell Carcinoma. Cancers 2018, 10, 443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.; Yang, N. MiR-335-5p Inhibits Cell Proliferation, Migration and Invasion in Colorectal Cancer through Downregulating LDHB. J. BUON Off. J. Balk. Union Oncol. 2019, 24, 1128–1136. [Google Scholar]
- Cui, J.; Quan, M.; Jiang, W.; Hu, H.; Jiao, F.; Li, N.; Jin, Z.; Wang, L.; Wang, Y.; Wang, L. Suppressed expression of LDHB promotes pancreatic cancer progression via inducing glycolytic phenotype. Med. Oncol. 2015, 32, 143. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.-H.; Kim, E.-L.; Lee, Y.-K.; Park, C.-B.; Kim, B.-W.; Wang, H.-J.; Yoon, C.-H.; Lee, S.-J.; Yoon, G. Decreased lactate dehydrogenase B expression enhances claudin 1-mediated hepatoma cell invasiveness via mitochondrial defects. Exp. Cell Res. 2011, 317, 1108–1118. [Google Scholar] [CrossRef]
- Frank, A.-C.; Raue, R.; Fuhrmann, D.C.; Sirait-Fischer, E.; Reuse, C.; Weigert, A.; Lütjohann, D.; Hiller, K.; Syed, S.N.; Brüne, B. Lactate dehydrogenase B regulates macrophage metabolism in the tumor microenvironment. Theranostics 2021, 11, 7570–7588. [Google Scholar] [CrossRef]
- Fu, D.; Li, J.; Wei, J.; Zhang, Z.; Luo, Y.; Tan, H.; Ren, C. HMGB2 is associated with malignancy and regulates Warburg effect by targeting LDHB and FBP1 in breast cancer. Cell Commun. Signal. CCS 2018, 16, 8. [Google Scholar] [CrossRef] [Green Version]
- Shibata, S.; Sogabe, S.; Miwa, M.; Fujimoto, T.; Takakura, N.; Naotsuka, A.; Kitamura, S.; Kawamoto, T.; Soga, T. Identification of the first highly selective inhibitor of human lactate dehydrogenase B. Sci. Rep. 2021, 11, 21353. [Google Scholar] [CrossRef]
- Monroe, G.R.; van Eerde, A.M.; Tessadori, F.; Duran, K.J.; Savelberg, S.M.C.; van Alfen, J.C.; Terhal, P.A.; van der Crabben, S.N.; Lichtenbelt, K.D.; Fuchs, S.A.; et al. Identification of human D lactate dehydrogenase deficiency. Nat. Commun. 2019, 10, 1477. [Google Scholar] [CrossRef] [Green Version]
- Drabkin, M.; Yogev, Y.; Zeller, L.; Zarivach, R.; Zalk, R.; Halperin, D.; Wormser, O.; Gurevich, E.; Landau, D.; Kadir, R.; et al. Hyperuricemia and gout caused by missense mutation in d-lactate dehydrogenase. J. Clin. Investig. 2019, 129, 5163–5168. [Google Scholar] [CrossRef] [Green Version]
- Song, K.-J.; Yu, X.-N.; Lv, T.; Chen, Y.-L.; Diao, Y.-C.; Liu, S.-L.; Wang, Y.-K.; Yao, Q. Expression and prognostic value of lactate dehydrogenase-A and -D subunits in human uterine myoma and uterine sarcoma. Medicine 2018, 97, e0268. [Google Scholar] [CrossRef]
- Olson, S.T.; Massey, V.; Ghisla, S.; Whitfield, C.D. Suicide inactivation of the flavoenzyme D-lactate dehydrogenase by alpha-hydroxybutynoate. Biochemistry 1979, 18, 4724–4732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yumnam, S.; Subedi, L.; Kim, S.Y. Glyoxalase System in the Progression of Skin Aging and Skin Malignancies. Int. J. Mol. Sci. 2020, 22, 310. [Google Scholar] [CrossRef] [PubMed]
- Kold-Christensen, R.; Johannsen, M. Methylglyoxal Metabolism and Aging-Related Disease: Moving from Correlation toward Causation. Trends Endocrinol. Metab. TEM 2020, 31, 81–92. [Google Scholar] [CrossRef]
- Rabbani, N.; Xue, M.; Weickert, M.O.; Thornalley, P.J. Multiple roles of glyoxalase 1-mediated suppression of methylglyoxal glycation in cancer biology-Involvement in tumour suppression, tumour growth, multidrug resistance and target for chemotherapy. Semin. Cancer Biol. 2018, 49, 83–93. [Google Scholar] [CrossRef] [PubMed]
- Thornalley, P.J.; Rabbani, N. Glyoxalase in tumourigenesis and multidrug resistance. Semin. Cell Dev. Biol. 2011, 22, 318–325. [Google Scholar] [CrossRef] [PubMed]
- Thornalley, P.J. Protecting the genome: Defence against nucleotide glycation and emerging role of glyoxalase I overexpression in multidrug resistance in cancer chemotherapy. Biochem. Soc. Trans. 2003, 31, 1372–1377. [Google Scholar] [CrossRef] [PubMed]
- Santarius, T.; Bignell, G.R.; Greenman, C.D.; Widaa, S.; Chen, L.; Mahoney, C.L.; Butler, A.; Edkins, S.; Waris, S.; Thornalley, P.J.; et al. GLO1-A novel amplified gene in human cancer. Genes Chromosomes Cancer 2010, 49, 711–725. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Zhang, C.; Zhang, L.; Yang, Q.; Zhou, S.; Wen, Q.; Wang, J. Nrf2 is a potential prognostic marker and promotes proliferation and invasion in human hepatocellular carcinoma. BMC Cancer 2015, 15, 531. [Google Scholar] [CrossRef] [Green Version]
- MacLeod, A.K.; Acosta-Jimenez, L.; Coates, P.J.; McMahon, M.; Carey, F.A.; Honda, T.; Henderson, C.J.; Wolf, C.R. Aldo-keto reductases are biomarkers of NRF2 activity and are co-ordinately overexpressed in non-small cell lung cancer. Br. J. Cancer 2016, 115, 1530–1539. [Google Scholar] [CrossRef] [Green Version]
- Romero, R.; Sayin, V.I.; Davidson, S.M.; Bauer, M.R.; Singh, S.X.; LeBoeuf, S.E.; Karakousi, T.R.; Ellis, D.C.; Bhutkar, A.; Sánchez-Rivera, F.J.; et al. Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis. Nat. Med. 2017, 23, 1362–1368. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.S.; Zhang, Z.G.; Du, G.Y.; Sun, H.L.; Liu, H.Y.; Zhou, Z.; Gou, X.M.; Wu, X.H.; Yu, X.Y.; Huang, Y.H. Nrf2 promotes breast cancer cell migration via up-regulation of G6PD/HIF-1alpha/Notch1 axis. J. Cell. Mol. Med. 2019, 23, 3451–3463. [Google Scholar] [CrossRef] [Green Version]
- Xue, M.; Rabbani, N.; Momiji, H.; Imbasi, P.; Anwar, M.M.; Kitteringham, N.; Park, B.K.; Souma, T.; Moriguchi, T.; Yamamoto, M.; et al. Transcriptional control of glyoxalase 1 by Nrf2 provides a stress-responsive defence against dicarbonyl glycation. Biochem. J. 2012, 443, 213–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rabbani, N.; Xue, M.; Thornalley, P.J. Activity, regulation, copy number and function in the glyoxalase system. Biochem. Soc. Trans. 2014, 42, 419–424. [Google Scholar] [CrossRef] [PubMed]
- Redon, R.; Ishikawa, S.; Fitch, K.R.; Feuk, L.; Perry, G.H.; Andrews, T.D.; Fiegler, H.; Shapero, M.H.; Carson, A.R.; Chen, W.; et al. Global variation in copy number in the human genome. Nature 2006, 444, 444–454. [Google Scholar] [CrossRef]
- Hutschenreuther, A.; Bigl, M.; Hemdan, N.Y.A.; Debebe, T.; Gaunitz, F.; Birkenmeier, G. Modulation of GLO1 Expression Affects Malignant Properties of Cells. Int. J. Mol. Sci. 2016, 17, 2133. [Google Scholar] [CrossRef] [Green Version]
- Guo, Y.; Zhang, Y.; Yang, X.; Lu, P.; Yan, X.; Xiao, F.; Zhou, H.; Wen, C.; Shi, M.; Lu, J.; et al. Effects of methylglyoxal and glyoxalase I inhibition on breast cancer cells proliferation, invasion, and apoptosis through modulation of MAPKs, MMP9, and Bcl-2. Cancer Biol. Ther. 2016, 17, 169–180. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Fang, L.; Li, G.; Zhang, J.; Li, C.; Ma, M.; Guan, C.; Bai, F.; Lyu, J.; Meng, Q.H. Synergistic inhibition of colon cancer growth by the combination of methylglyoxal and silencing of glyoxalase I mediated by the STAT1 pathway. Oncotarget 2017, 8, 54838–54857. [Google Scholar] [CrossRef] [Green Version]
- Jin, T.; Zhao, L.; Wang, H.-P.; Huang, M.-L.; Yue, Y.; Lu, C.; Zheng, Z.B. Recent advances in the discovery and development of glyoxalase I inhibitors. Bioorg. Med. Chem. 2020, 28, 115243. [Google Scholar] [CrossRef]
- Zender, L.; Xue, W.; Zuber, J.; Semighini, C.P.; Krasnitz, A.; Ma, B.; Zender, P.; Kubicka, S.; Luk, J.M.; Schirmacher, P.; et al. An oncogenomics-based in vivo RNAi screen identifies tumor suppressors in liver cancer. Cell 2008, 135, 852–864. [Google Scholar] [CrossRef] [Green Version]
- Xue, M.; Weickert, M.O.; Qureshi, S.; Kandala, N.B.; Anwar, A.; Waldron, M.; Shafie, A.; Messenger, D.; Fowler, M.; Jenkins, G.; et al. Improved Glycemic Control and Vascular Function in Overweight and Obese Subjects by Glyoxalase 1 Inducer Formulation. Diabetes 2016, 65, 2282–2294. [Google Scholar] [CrossRef] [Green Version]
- Talesa, V.N.; Ferri, I.; Bellezza, G.; Love, H.D.; Sidoni, A.; Antognelli, C. Glyoxalase 2 Is Involved in Human Prostate Cancer Progression as Part of a Mechanism Driven By PTEN/PI3K/AKT/mTOR Signaling With Involvement of PKM2 and ERalpha. Prostate 2017, 77, 196–210. [Google Scholar] [CrossRef]
- Antognelli, C.; Ferri, I.; Bellezza, G.; Siccu, P.; Love, H.D.; Talesa, V.N.; Sidoni, A. Glyoxalase 2 drives tumorigenesis in human prostate cells in a mechanism involving androgen receptor and p53-p21 axis. Mol. Carcinog. 2017, 56, 2112–2126. [Google Scholar] [CrossRef]
- Halestrap, A.P. The SLC16 gene family—Structure, role and regulation in health and disease. Mol. Asp. Med. 2013, 34, 337–349. [Google Scholar] [CrossRef] [PubMed]
- Halestrap, A.P.; Meredith, D. The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflug. Arch. Eur. J. Physiol. 2004, 447, 619–628. [Google Scholar] [CrossRef] [PubMed]
- Halestrap, A.P.; Wilson, M.C. The monocarboxylate transporter family--role and regulation. IUBMB Life 2012, 64, 109–119. [Google Scholar] [CrossRef] [PubMed]
- Halestrap, A.P. Monocarboxylic acid transport. Compr. Physiol. 2013, 3, 1611–1643. [Google Scholar]
- Payen, V.L.; Mina, E.; Van Hee, V.F.; Porporato, P.E.; Sonveaux, P. Monocarboxylate transporters in cancer. Mol. Metab. 2020, 33, 48–66. [Google Scholar] [CrossRef]
- Sonveaux, P.; Vegran, F.; Schroeder, T.; Wergin, M.C.; Verrax, J.; Rabbani, Z.N. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Investig. 2008, 118, 3930–3942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Whitaker-Menezes, D.; Martinez-Outschoorn, U.E.; Lin, Z.; Ertel, A.; Flomenberg, N.; Witkiewicz, A.K.; Birbe, R.; Howell, A.; Pavlides, S.; Lisanti, M.P.; et al. Evidence for a stromal-epithelial “lactate shuttle” in human tumors: MCT4 is a marker of oxidative stress in cancer-associated fibroblasts. Cell cycle 2011, 10, 1772–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pinheiro, C.; Reis, R.M.; Ricardo, S.; Longatto-Filho, A.; Schmitt, F.; Baltazar, F. Expression of monocarboxylate transporters 1, 2, and 4 in human tumours and their association with CD147 and CD44. J. Bio. Biotechnol. 2010, 2010, 427694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Garcia-Canaveras, J.C.; Chen, L.; Rabinowitz, J.D. The Tumor Metabolic Microenvironment: Lessons from Lactate. Cancer Res. 2019, 79, 3155–3162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kroemer, G.; Pouyssegur, J. Tumor cell metabolism: Cancer’s Achilles’ heel. Cancer Cell 2008, 13, 472–482. [Google Scholar] [CrossRef] [PubMed]
- Semenza, G.L. Tumor metabolism: Cancer cells give and take lactate. J. Clin. Investig. 2008, 118, 3835–3837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, X.; Wang, M.; Wang, M.; Yao, L.; Li, X.; Dong, H.; Li, M.; Sun, T.; Liu, X.; Liu, Y.; et al. Role of Proton-Coupled Monocarboxylate Transporters in Cancer: From Metabolic Crosstalk to Therapeutic Potential. Front. Cell Dev. Biol. 2020, 8, 651. [Google Scholar] [CrossRef] [PubMed]
- Rohlenova, K.; Veys, K.; Miranda-Santos, I.; De Bock, K.; Carmeliet, P. Endothelial Cell Metabolism in Health and Disease. Trends. Cell Biol. 2018, 28, 224–236. [Google Scholar] [CrossRef]
- Guo, C.; Huang, T.; Wang, Q.H.; Li, H.; Khanal, A.; Kang, E.H. Monocarboxylate transporter 1 and monocarboxylate transporter 4 in cancer-endothelial co-culturing microenvironments promote proliferation, migration, and invasion of renal cancer cells. Cancer Cell Int. 2019, 19, 170. [Google Scholar] [CrossRef]
- Vegran, F.; Boidot, R.; Michiels, C.; Sonveaux, P.; Feron, O. Lactate influx through the endothelial cell monocarboxylate transporter MCT1 supports an NF-kappaB/IL-8 pathway that drives tumor angiogenesis. Cancer Res. 2011, 71, 2550–2560. [Google Scholar] [CrossRef] [Green Version]
- Miranda-Goncalves, V.; Bezerra, F.; Costa-Almeida, R.; Freitas-Cunha, M.; Soares, R.; Martinho, O. Monocarboxylate transporter 1 is a key player in glioma-endothelial cell crosstalk. Mol. Carcinog. 2017, 56, 2630–2642. [Google Scholar] [CrossRef]
- Alves, V.A.; Pinheiro, C.; Morais-Santos, F.; Felipe-Silva, A.; Longatto-Filho, A.; Baltazar, F. Characterization of monocarboxylate transporter activity in hepatocellular carcinoma. World J. Gastroenterol. 2014, 20, 11780–11787. [Google Scholar] [CrossRef] [Green Version]
- Curtis, N.J.; Mooney, L.; Hopcroft, L.; Michopoulos, F.; Whalley, N.; Zhong, H. Pre-clinical pharmacology of AZD3965, a selective inhibitor of MCT1: DLBCL, NHL and Burkitt’s lymphoma anti-tumor activity. Oncotarget 2017, 8, 69219–69236. [Google Scholar] [CrossRef] [Green Version]
- Noble, R.A.; Bell, N.; Blair, H.; Sikka, A.; Thomas, H.; Phillips, N.; Nakjang, S.; Miwa, S.; Crossland, R.; Rand, V.; et al. Inhibition of monocarboxyate transporter 1 by AZD3965 as a novel therapeutic approach for diffuse large B-cell lymphoma and Burkitt lymphoma. Haematologica 2017, 102, 1247–1257. [Google Scholar] [CrossRef]
- Beloueche-Babari, M.; Wantuch, S.; Casals Galobart, T.; Koniordou, M.; Parkes, H.G.; Arunan, V. MCT1 Inhibitor AZD3965 Increases Mitochondrial Metabolism, Facilitating Combination Therapy and Noninvasive Magnetic Resonance Spectroscopy. Cancer Res. 2017, 77, 5913–5924. [Google Scholar] [CrossRef] [Green Version]
- Doherty, J.R.; Yang, C.; Scott, K.E.; Cameron, M.D.; Fallahi, M.; Li, W.; Hall, A.; Amelio, A.L.; Mishra, J.K.; Li, F.; et al. Blocking lactate export by inhibiting the Myc target MCT1 Disables glycolysis and glutathione synthesis. Cancer Res. 2014, 74, 908–920. [Google Scholar] [CrossRef] [Green Version]
- Murray, C.M.; Hutchinson, R.; Bantick, J.R.; Belfield, G.P.; Benjamin, A.D.; Brazma, D.; Bundick, V.; Cook, I.D.; Craggs, R.I.; Edwards, S.; et al. Monocarboxylate transporter MCT1 is a target for immunosuppression. Nat. Chem. Biol. 2005, 1, 371–376. [Google Scholar] [CrossRef]
- Beloueche-Babari, M.; Casals Galobart, T.; Delgado-Goni, T.; Wantuch, S.; Parkes, H.G.; Tandy, D.; Harker, J.A.; Leach, M.O. Monocarboxylate transporter 1 blockade with AZD3965 inhibits lipid biosynthesis and increases tumour immune cell infiltration. Br. J. Cancer 2020, 122, 895–903. [Google Scholar] [CrossRef]
- Wang, K.; Huang, W.; Chen, R.; Lin, P.; Zhang, T.; Ni, Y.-F.; Li, H.; Wu, J.; Sun, X.-X.; Geng, J.-J.; et al. Di-methylation of CD147-K234 Promotes the Progression of NSCLC by Enhancing Lactate Export. Cell Metab. 2021, 33, 160–173.e6. [Google Scholar] [CrossRef]
- Ullah, M.S.; Davies, A.J.; Halestrap, A.P. The plasma membrane lactate transporter MCT4, but not MCT1, is up-regulated by hypoxia through a HIF-1alpha-dependent mechanism. J. Biol. Chem. 2006, 281, 9030–9037. [Google Scholar] [CrossRef] [Green Version]
- Ahmed, K. Biological roles and therapeutic potential of hydroxy-carboxylic Acid receptors. Front. Endocrinol. 2011, 2, 51. [Google Scholar] [CrossRef] [Green Version]
- Ge, H.; Weiszmann, J.; Reagan, J.D.; Gupte, J.; Baribault, H.; Gyuris, T.; Chen, J.-L.; Tian, H.; Li, Y. Elucidation of signaling and functional activities of an orphan GPCR, GPR81. J. Lipid Res. 2008, 49, 797–803. [Google Scholar] [CrossRef] [Green Version]
- Cai, T.-Q.; Ren, N.; Jin, L.; Cheng, K.; Kash, S.; Chen, R.; Wright, S.D.; Taggart, A.K.; Waters, M.G. Role of GPR81 in lactate-mediated reduction of adipose lipolysis. Biochem. Biophys. Res. Commun. 2008, 377, 987–991. [Google Scholar] [CrossRef]
- Liu, C.; Wu, J.; Zhu, J.; Kuei, C.; Yu, J.; Shelton, J.; Lovenberg, T.W. Lactate inhibits lipolysis in fat cells through activation of an orphan G-protein-coupled receptor, GPR81. J. Biol. Chem. 2009, 284, 2811–2822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, T.P.; Ganapathy, V. Lactate/GPR81 signaling and proton motive force in cancer: Role in angiogenesis, immune escape, nutrition, and Warburg phenomenon. Pharmacol. Ther. 2020, 206, 107451. [Google Scholar] [CrossRef] [PubMed]
- Xie, Q.; Zhu, Z.; He, Y.; Zhang, Z.; Zhang, Y.; Wang, Y.; Luo, J.; Peng, T.; Cheng, F.; Gao, J.; et al. A lactate-induced Snail/STAT3 pathway drives GPR81 expression in lung cancer cells. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165576. [Google Scholar] [CrossRef] [PubMed]
- Lee, D.K.; Nguyen, T.; Lynch, K.R.; Cheng, R.; Vanti, W.B.; Arkhitko, O.; George, S.R.; O’Dowd, B.F. Discovery and mapping of ten novel G protein-coupled receptor genes. Gene 2001, 275, 83–91. [Google Scholar] [CrossRef]
- Ippolito, L.; Marini, A.; Cavallini, L.; Morandi, A.; Pietrovito, L.; Pintus, G.; Giannoni, E.; Schrader, T.; Puhr, M.; Chiarugi, P.; et al. Metabolic shift toward oxidative phosphorylation in docetaxel resistant prostate cancer cells. Oncotarget 2016, 7, 61890–61904. [Google Scholar] [CrossRef]
- Viale, A.; Pettazzoni, P.; Lyssiotis, C.A.; Ying, H.; Sanchez, N.; Marchesini, M. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 2014, 514, 628–632. [Google Scholar] [CrossRef] [Green Version]
- Hu, J.; Cai, M.; Liu, Y.; Liu, B.; Xue, X.; Ji, R.; Bian, X.; Lou, S. The roles of GRP81 as a metabolic sensor and inflammatory mediator. J. Cell. Physiol. 2020, 235, 8938–8950. [Google Scholar] [CrossRef]
- Zohn, I.E.; Klinger, M.; Karp, X.; Kirk, H.; Symons, M.; Chrzanowska-Wodnicka, M.; Der, C.J.; Kay, R.J. G2A is an oncogenic G protein-coupled receptor. Oncogene 2000, 19, 3866–3877. [Google Scholar] [CrossRef] [Green Version]
- Cheng, W.Y.; 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. [Google Scholar] [CrossRef]
- Bolick, D.T.; Skaflen, M.D.; Johnson, L.E.; Kwon, S.-C.; Howatt, D.; Daugherty, A.; Ravichandran, K.S.; Hedrick, C.C. G2A deficiency in mice promotes macrophage activation and atherosclerosis. Circ. Res. 2009, 104, 318–327. [Google Scholar] [CrossRef] [Green Version]
- Justus, C.R.; Dong, L.; Yang, L.V. Acidic tumor microenvironment and pH-sensing G protein-coupled receptors. Front. Physiol. 2013, 4, 354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Radu, C.G.; Yang, L.V.; Riedinger, M.; Au, M.; Witte, O.N. T cell chemotaxis to lysophosphatidylcholine through the G2A receptor. Proc. Natl. Acad. Sci. USA 2004, 101, 245–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kabarowski, J.H. G2A and LPC: Regulatory functions in immunity. Prostaglandins Other Lipid Mediat. 2009, 89, 73–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schenz, J.; Heilig, L.; Lohse, T.; Tichy, L.; Bomans, K.; Buttner, M.; Weigand, M.A.; Uhle, F. Extracellular Lactate Acts as a Metabolic Checkpoint and Shapes Monocyte Function Time Dependently. Front. Immunol. 2021, 12, 729209. [Google Scholar] [CrossRef] [PubMed]
- Caslin, H.L.; Abebayehu, D.; Pinette, J.A.; Ryan, J.J. Lactate Is a Metabolic Mediator That Shapes Immune Cell Fate and Function. Front. Physiol. 2021, 12, 688485. [Google Scholar] [CrossRef] [PubMed]
- Murakami, N.; Yokomizo, T.; Okuno, T.; Shimizu, T. G2A is a proton-sensing G-protein-coupled receptor antagonized by lysophosphatidylcholine. J. Biol. Chem. 2004, 279, 42484–42491. [Google Scholar] [CrossRef] [Green Version]
- Brooks, G.A. Cell-cell and intracellular lactate shuttles. J. Physiol. 2009, 587, 5591–5600. [Google Scholar] [CrossRef]
- Peluso, I.; Yarla, N.S.; Ambra, R.; Pastore, G.; Perry, G. MAPK signalling pathway in cancers: Olive products as cancer preventive and therapeutic agents. Semin. Cancer Biol. 2019, 56, 185–195. [Google Scholar] [CrossRef]
- Lee, S.; Rauch, J.; Kolch, W. Targeting MAPK Signaling in Cancer: Mechanisms of Drug Resistance and Sensitivity. Int. J. Mol. Sci. 2020, 21, 1102. [Google Scholar] [CrossRef] [Green Version]
- Mu, X.; Shi, W.; Xu, Y.; Xu, C.; Zhao, T.; Geng, B.; Pan, J.; Hu, S.; Zhang, C.; Zhang, J.; et al. Tumor-derived lactate induces M2 macrophage polarization via the activation of the ERK/STAT3 signaling pathway in breast cancer. Cell Cycle 2018, 17, 428–438. [Google Scholar] [CrossRef] [Green Version]
- Albadari, N.; Deng, S.; Li, W. The transcriptional factors HIF-1 and HIF-2 and their novel inhibitors in cancer therapy. Expert Opin. Drug Discov. 2019, 14, 667–682. [Google Scholar] [CrossRef] [PubMed]
- Bredholt, G.; Mannelqvist, M.; Stefansson, I.M.; Birkeland, E.; Bø, T.H.; Øyan, A.M.; Trovik, J.; Kalland, K.-H.; Jonassen, I.; Salvesen, H.B.; et al. Tumor necrosis is an important hallmark of aggressive endometrial cancer and associates with hypoxia, angiogenesis and inflammation responses. Oncotarget 2015, 6, 39676–39691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Multhoff, G.; Vaupel, P. Hypoxia Compromises Anti-Cancer Immune Responses. Adv. Exp. Med. Biol. 2020, 1232, 131–143. [Google Scholar] [PubMed]
- Wang, M.; Zhao, X.; Zhu, D.; Liu, T.; Liang, X.; Liu, F.; Sun, B. HIF-1alpha promoted vasculogenic mimicry formation in hepatocellular carcinoma through LOXL2 up-regulation in hypoxic tumor microenvironment. J. Exp. Clin. Cancer Res. 2017, 36, 60. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Yan, W.; Tohme, S.; Chen, M.; Fu, Y.; Tian, D.; Lotze, M.; Tang, D.; Tsung, A. Hypoxia induced HMGB1 and mitochondrial DNA interactions mediate tumor growth in hepatocellular carcinoma through Toll-like receptor 9. J. Hepatol. 2015, 63, 114–121. [Google Scholar] [CrossRef]
- de Heer, E.C.; Jalving, M.; Harris, A.L. HIFs, angiogenesis, and metabolism: Elusive enemies in breast cancer. J. Clin. Investig. 2020, 130, 5074–5087. [Google Scholar] [CrossRef]
- De Peaux, K.; Delgoffe, G.M. Metabolic barriers to cancer immunotherapy. Nat. Rev. Immunol. 2021, 21, 785–797. [Google Scholar] [CrossRef]
- Riera-Domingo, C.; Audige, A.; Granja, S.; Cheng, W.C.; Ho, P.C.; Baltazar, F.; Mazzone, M. Immunity, Hypoxia, and Metabolism-the Menage a Trois of Cancer: Implications for Immunotherapy. Physiol. Rev. 2020, 100, 1–102. [Google Scholar] [CrossRef]
- Semenza, G.L. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J. Clin. Investig. 2013, 123, 3664–3671. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Lu, H.; Chen, C.; Lyu, Y.; Cole, R.N.; Semenza, G.L. HIF-1 Interacts with TRIM28 and DNA-PK to release paused RNA polymerase II and activate target gene transcription in response to hypoxia. Nat. Commun. 2022, 13, 316. [Google Scholar] [CrossRef]
- Icard, P.; Shulman, S.; Farhat, D.; Steyaert, J.M.; Alifano, M.; Lincet, H. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist. Updates 2018, 38, 1–11. [Google Scholar] [CrossRef] [PubMed]
- McDonald, P.C.; Chafe, S.C.; Brown, W.S.; Saberi, S.; Swayampakula, M.; Venkateswaran, G.; Nemirovsky, O.; Gillespie, J.A.; Karasinska, J.M.; Kalloger, S.E.; et al. Regulation of pH by Carbonic Anhydrase 9 Mediates Survival of Pancreatic Cancer Cells With Activated KRAS in Response to Hypoxia. Gastroenterology 2019, 157, 823–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ivashkiv, L.B. The hypoxia-lactate axis tempers inflammation. Nat. Rev. Immunol. 2020, 20, 85–86. [Google Scholar] [CrossRef] [PubMed]
- Sonveaux, P.; Copetti, T.; De Saedeleer, C.J.; Vegran, F.; Verrax, J.; Kennedy, K.M.; Moon, E.J.; Dhup, S.; Danhier, P.; Frérart, F.; et al. Targeting the lactate transporter MCT1 in endothelial cells inhibits lactate-induced HIF-1 activation and tumor angiogenesis. PLoS ONE 2012, 7, e33418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Saedeleer, C.J.; Copetti, T.; Porporato, P.E.; Verrax, J.; Feron, O.; Sonveaux, P. Lactate activates HIF-1 in oxidative but not in Warburg-phenotype human tumor cells. PLoS ONE 2012, 7, e46571. [Google Scholar] [CrossRef]
- Ippolito, L.; Morandi, A.; Taddei, M.L.; Parri, M.; Comito, G.; Iscaro, A. Cancer-associated fibroblasts promote prostate cancer malignancy via metabolic rewiring and mitochondrial transfer. Oncogene 2019, 38, 5339–5355. [Google Scholar] [CrossRef]
- Park, K.C.; Lee, D.C.; Yeom, Y.I. NDRG3-mediated lactate signaling in hypoxia. BMB Rep. 2015, 48, 301–302. [Google Scholar] [CrossRef] [Green Version]
- Yao, Y.; Wang, W.; Jing, L.; Wang, Y.; Li, M.; Hou, X.; Wang, J.; Peng, T.; Teng, J.; Jia, Y. Let-7f Regulates the Hypoxic Response in Cerebral Ischemia by Targeting NDRG3. Neurochem. Res. 2017, 42, 446–454. [Google Scholar] [CrossRef]
- Guerrero-Zotano, A.; Mayer, I.A.; Arteaga, C.L. PI3K/AKT/mTOR: Role in breast cancer progression, drug resistance, and treatment. Cancer Metastasis Rev. 2016, 35, 515–524. [Google Scholar] [CrossRef]
- Ediriweera, M.K.; Tennekoon, K.H.; Samarakoon, S.R. Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: Biological and therapeutic significance. Semin. Cancer Biol. 2019, 59, 147–160. [Google Scholar] [CrossRef]
- Chen, H.; Zhou, L.; Wu, X.; Li, R.; Wen, J.; Sha, J.; Wen, X. The PI3K/AKT pathway in the pathogenesis of prostate cancer. Front. Biosci. -Landmark 2016, 21, 1084–1091. [Google Scholar]
- Noorolyai, S.; Shajari, N.; Baghbani, E.; Sadreddini, S.; Baradaran, B. The relation between PI3K/AKT signalling pathway and cancer. Gene 2019, 698, 120–128. [Google Scholar] [CrossRef] [PubMed]
- Courtnay, R.; Ngo, D.C.; Malik, N.; Ververis, K.; Tortorella, S.M.; Karagiannis, T.C. Cancer metabolism and the Warburg effect: The role of HIF-1 and PI3K. Mol. Biol. Rep. 2015, 42, 841–851. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hennessy, B.T.; Smith, D.L.; Ram, P.T.; Lu, Y.; Mills, G.B. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug Discov. 2005, 4, 988–1004. [Google Scholar] [CrossRef]
- Fresno Vara, J.A.; Casado, E.; de Castro, J.; Cejas, P.; Belda-Iniesta, C.; Gonzalez-Baron, M. PI3K/Akt signalling pathway and cancer. Cancer Treat. Rev. 2004, 30, 193–204. [Google Scholar] [CrossRef]
- Dolcet, X.; Llobet, D.; Pallares, J.; Matias-Guiu, X. NF-kB in development and progression of human cancer. Virchows Arch. 2005, 446, 475–482. [Google Scholar] [CrossRef]
- Lehman, H.L.; Kidacki, M.; Warrick, J.I.; Stairs, D.B. NFkB hyperactivation causes invasion of esophageal squamous cell carcinoma with EGFR overexpression and p120-catenin down-regulation. Oncotarget 2018, 9, 11180–11196. [Google Scholar] [CrossRef] [Green Version]
- Jana, A.; Krett, N.L.; Guzman, G.; Khalid, A.; Ozden, O.; Staudacher, J.J. NFkB is essential for activin-induced colorectal cancer migration via upregulation of PI3K-MDM2 pathway. Oncotarget 2017, 8, 37377–37393. [Google Scholar] [CrossRef] [Green Version]
- Chang, C.P.; Su, Y.C.; Lee, P.H.; Lei, H.Y. Targeting NFKB by autophagy to polarize hepatoma-associated macrophage differentiation. Autophagy 2013, 9, 619–621. [Google Scholar] [CrossRef] [Green Version]
- Dan, H.; Liu, S.; Liu, J.; Liu, D.; Yin, F.; Wei, Z. RACK1 promotes cancer progression by increasing the M2/M1 macrophage ratio via the NF-kappaB pathway in oral squamous cell carcinoma. Mol. Oncol. 2020, 14, 795–807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, X.; Zhang, M.; Xu, F.; Jiang, S. Wnt signaling in breast cancer: Biological mechanisms, challenges and opportunities. Mol. Cancer 2020, 19, 165. [Google Scholar] [CrossRef] [PubMed]
- Parsons, M.J.; Tammela, T.; Dow, L.E. WNT as a Driver and Dependency in Cancer. Cancer Discov. 2021, 11, 2413–2429. [Google Scholar] [CrossRef] [PubMed]
- Krishnamurthy, N.; Kurzrock, R. Targeting the Wnt/beta-catenin pathway in cancer: Update on effectors and inhibitors. Cancer Treat. Rev. 2018, 62, 50–60. [Google Scholar] [CrossRef]
- Zhan, T.; Rindtorff, N.; Boutros, M. Wnt signaling in cancer. Oncogene 2017, 36, 1461–1473. [Google Scholar] [CrossRef]
- Reya, T.; Clevers, H. Wnt signalling in stem cells and cancer. Nature 2005, 434, 843–850. [Google Scholar] [CrossRef]
- Lee, Y.-S.; Kim, T.-Y.; Kim, Y.; Lee, S.-H.; Kim, S.; Kang, S.W. Microbiota-Derived Lactate Accelerates Intestinal Stem-Cell-Mediated Epithelial Development. Cell Host Microbe 2018, 24, 833–846.e6. [Google Scholar] [CrossRef] [Green Version]
- Madaan, A.; Chaudhari, P.; Nadeau-Vallée, M.; Hamel, D.; Zhu, T.; Mitchell, G. Müller Cell–Localized G-Protein–Coupled Receptor 81 (Hydroxycarboxylic Acid Receptor 1) Regulates Inner Retinal Vasculature via Norrin/Wnt Pathways. Am. J. Pathol. 2019, 189, 1878–1896. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cai, M.; Wan, J.; Cai, K.; Song, H.; Wang, Y.; Sun, W.; Hu, J. Understanding the Contribution of Lactate Metabolism in Cancer Progress: A Perspective from Isomers. Cancers 2023, 15, 87. https://doi.org/10.3390/cancers15010087
Cai M, Wan J, Cai K, Song H, Wang Y, Sun W, Hu J. Understanding the Contribution of Lactate Metabolism in Cancer Progress: A Perspective from Isomers. Cancers. 2023; 15(1):87. https://doi.org/10.3390/cancers15010087
Chicago/Turabian StyleCai, Ming, Jian Wan, Keren Cai, Haihan Song, Yujiao Wang, Wanju Sun, and Jingyun Hu. 2023. "Understanding the Contribution of Lactate Metabolism in Cancer Progress: A Perspective from Isomers" Cancers 15, no. 1: 87. https://doi.org/10.3390/cancers15010087
APA StyleCai, M., Wan, J., Cai, K., Song, H., Wang, Y., Sun, W., & Hu, J. (2023). Understanding the Contribution of Lactate Metabolism in Cancer Progress: A Perspective from Isomers. Cancers, 15(1), 87. https://doi.org/10.3390/cancers15010087