We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

GLUT1 biological function and inhibition: research advances

    Shuang Cao‡

    Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yongshou Chen‡

    Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Yixin Ren

    Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China

    ,
    Yingqi Feng

    **Author for correspondence:

    E-mail Address: fengyingqivip@163.com

    Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China

    &
    Sihui Long

    *Author for correspondence:

    E-mail Address: sihuilong@wit.edu.cn

    Key Laboratory of Green Chemical Engineering Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, PR China

    Published Online:https://doi.org/10.4155/fmc-2021-0071

    The GLUT is a key regulator of glucose metabolism and is widely expressed on the surface of most cells of the body. GLUT provides a variety of nutrients for the growth, proliferation and differentiation of cells. In recent years, the development of drugs affecting the energy intake of tumor cells has become a research hotspot. GLUT inhibitors are gaining increased attention because they can block the energy supply of malignant tumors. Herein, we elaborate on the structure and function of GLUT1, the structural and functional differences among GLUT1–4 transporters and the relationship between GLUT1 and tumor development, as well as GLUT1 transporter inhibitors, to provide a reference for the development of new GLUT1 inhibitors.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Ferreira LM. Cancer metabolism: the Warburg effect today. Exp. Mol. Pathol. 89(3), 372–380 (2010).
    • 2. Abdou AG, Eldien MM, Elsakka D. GLUT-1 expression in cutaneous basal and squamous cell carcinomas. Int. J. Surg. Pathol. 23(6), 447–453 (2015).
    • 3. Ancey PB, Contat C, Meylan E. Glucose transporters in cancer – from tumor cells to the tumor microenvironment. FEBS. J. 285(16), 2926–2943 (2018).
    • 4. Ma X, Hui Y, Lin L et al. Clinical significance of COX-2, GLUT-1 and VEGF expressions in endometrial cancer tissues. Pak. J. Med. Sci. 31(2), 280–284 (2015).
    • 5. Qamar S, Fatima S, Rehman A et al. Glucose transporter 1 overexpression in oral squamous cell carcinoma. J. Coll. Physicians Surg. Pak. 29(8), 724–727 (2019).
    • 6. Mueckler M, Thorens B. The SLC2 (GLUT) family of membrane transporters. Mol. Aspects Med. 34(2–3), 121–138 (2013).
    • 7. Carruthers A, Dezutter J, Ganguly A, Devaskar SU. Will the original glucose transporter isoform please stand up. Am. J. Physiol. Endocrinol. Metab. 297(4), E836–848 (2009).
    • 8. Uldry M, Ibberson M, Hosokawa M, Thorens B. GLUT2 is a high affinity glucosamine transporter. FEBS Lett. 524(1–3), 199–203 (2002).
    • 9. Reckzeh ES, Waldmann H. Development of glucose transporter (GLUT) inhibitors. Eur. J. Org. Chem. 2020(16), 2321–2329 (2020).
    • 10. Uldry M, Thorens B. The SLC2 family of facilitated hexose and polyol transporters. Pflugers. Arch. 447(5), 480–489 (2004).
    • 11. Joost HG, Thorens B. The extended GLUT-family of sugar/polyol transport facilitators: nomenclature, sequence characteristics, and potential function of its novel members (review). Mol. Membr. Biol. 18(4), 247–256 (2001).
    • 12. Caulfield MJ, Munroe PB, O'Neill D et al. SLC2A9 is a high-capacity urate transporter in humans. PLoS Med. 5(10), e197 (2008).
    • 13. Deng D, Xu C, Sun P et al. Crystal structure of the human glucose transporter GLUT1. Nature 510(7503), 121–125 (2014). •• In this paper, the 3D crystal structure of GLUT1 was reported for the first time, and the structure and transport mechanism of GLUT1 were described in detail.
    • 14. Graybill C, Van Hoek AN, Desai D et al. Ultrastructure of human erythrocyte GLUT1. Biochemistry 45(26), 8096–8107 (2006).
    • 15. Zottola RJ, Cloherty EK, Coderre PE et al. Glucose transporter function is controlled by transporter oligomeric structure. A single, intramolecular disulfide promotes GLUT1 tetramerization. Biochemistry 34(30), 9734–9747 (1995).
    • 16. De Zutter JK, Levine KB, Deng D, Carruthers A. Sequence determinants of GLUT1 oligomerization: analysis by homology-scanning mutagenesis. J. Biol. Chem. 288(28), 20734–20744 (2013).
    • 17. Lloyd KP, Ojelabi OA, De Zutter JK, Carruthers A. Reconciling contradictory findings: glucose transporter 1 (GLUT1) functions as an oligomer of allosteric, alternating access transporters. J. Biol. Chem. 292(51), 21035–21046 (2017).
    • 18. Hamill S, Cloherty EK, Carruthers A. The human erythrocyte sugar transporter presents two sugar import sites. Biochemistry 38(51), 16974–16983 (1999).
    • 19. Baker GF, Naftalin RJ. Evidence of multiple operational affinities for D-glucose inside the human erythrocyte membrane. Biochim. Biophys. Acta. 550(3), 474–484 (1979).
    • 20. Naftalin RJ. Reassessment of models of facilitated transport and cotransport. J. Membr. Biol. 234(2), 75–112 (2010).
    • 21. Tian Y, Shen S, Gu L et al. Computer-aided design of glucoside brain-targeted molecules based on 4PYP. J. Mol. Graph. Model 103, 107819 (2020).
    • 22. Simpson IA, Dwyer D, Malide D et al. The facilitative glucose transporter GLUT3: 20 years of distinction. Am. J. Physiol-Endoc. M. 295(2), E242–E253 (2008).
    • 23. Huang S, Czech MP. The GLUT4 glucose transporter. Cell Metab. 5(4), 237–252 (2007).
    • 24. Chen C, Pore N, Behrooz A et al. Regulation of glut1 mRNA by hypoxia-inducible factor-1. Interaction between H-ras and hypoxia. J. Biol. Chem. 276(12), 9519–9525 (2001). • This paper describes the regulatory mechanism of HIF-1 on GLUT1 in detail.
    • 25. Weinstein SP, Haber RS. Glucose transport stimulation by thyroid hormone in ARL 15 cells: partial role of increased GLUT1 glucose transporter gene transcription. Thyroid 3(2), 135–142 (1993).
    • 26. Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res. 64(7), 2627–2633 (2004).
    • 27. Cifuentes M, Garcia MA, Arrabal PM et al. Insulin regulates GLUT1-mediated glucose transport in MG-63 human osteosarcoma cells. J. Cell Physiol. 226(6), 1425–1432 (2011).
    • 28. Tan VP, Miyamoto S. HK2/hexokinase-II integrates glycolysis and autophagy to confer cellular protection. Autophagy 11(6), 963–964 (2015).
    • 29. Angira D, Natarajan N, Dedania SR et al. Characterization of P. aeruginosa glucose 6-phosphate isomerase: a functional insight via in-vitro activity study. Curr. Top. Med. Chem. 20(29), 2651–2661 (2020).
    • 30. Webb BA, Forouhar F, Szu FE et al. Structures of human phosphofructokinase-1 and atomic basis of cancer-associated mutations. Nature 523(7558), 111–114 (2015).
    • 31. White MR, Garcin ED. D-glyceraldehyde-3-phosphate dehydrogenase structure and function. Sub-Cell. Biochem. 83, 413–453 (2017).
    • 32. Fu Q, Yu Z. Phosphoglycerate kinase 1 (PGK1) in cancer: a promising target for diagnosis and therapy. Life Sci. 256, 117863 (2020).
    • 33. Sharif F, Rasul A, Ashraf A et al. Phosphoglycerate mutase 1 in cancer: a promising target for diagnosis and therapy. IUBMB Life 71(10), 1418–1427 (2019).
    • 34. Counihan JL, Grossman EA, Nomura DK. Cancer metabolism: current understanding and therapies. Chem. Rev. 118(14), 6893–6923 (2018). • This paper describes the regulatory pathway of GLUT1.
    • 35. Ashton TM, McKenna WG, Kunz-Schughart LA, Higgins GS. Oxidative phosphorylation as an emerging target in cancer therapy. Clin. Cancer Res. 24(11), 2482–2490 (2018).
    • 36. Reckzeh ES, Waldmann H. Small-molecule inhibition of glucose transporters GLUT-1–4. Chembiochem 21(1), 45–52 (2020).
    • 37. Yin C, Gao B, Yang J, Wu J. Glucose transporter-1 (GLUT-1) expression is associated with tumor size and poor prognosis in locally advanced gastric cancer. Med. Sci. Monit. Basic Res. 26, e920778 (2020).
    • 38. Zhao M, Zhang Z. Glucose transporter regulation in cancer: a profile and the loops. Crit. Rev. Eukaryot Gene. Expr. 26(3), 223–238 (2016).
    • 39. Zhou JC, Zhang JJ, Zhang W et al. Expression of GLUT-1 in nasopharyngeal carcinoma and its clinical significance. Eur. Rev. Med. Pharmacol. Sci. 21(21), 4891–4895 (2017).
    • 40. Zhou L, Li S, Liu L et al. Recombinant methioninase regulates PI3K/Akt/Glut-1 pathway and inhibits aerobic glycolysis to promote apoptosis of gastric cancer cells. Nan Fang Yi Ke Da Xue Xue Bao 40(1), 27–33 (2020).
    • 41. National Cancer Institute. Cancer Stat Facts 2019. (2020). https://seer.cancer.gov/statfacts/html/corp.html
    • 42. Rudlowski C, Moser M, Becker AJ et al. GLUT1 mRNA and protein expression in ovarian borderline tumors and cancer. Oncology 66(5), 404–410 (2004).
    • 43. Xintaropoulou C, Ward C, Wise A et al. Expression of glycolytic enzymes in ovarian cancers and evaluation of the glycolytic pathway as a strategy for ovarian cancer treatment. BMC Cancer 18(1), 636 (2018).
    • 44. Ozcan A, Shen SS, Zhai QJ, Truong LD. Expression of GLUT1 in primary renal tumors: morphologic and biologic implications. Am. J. Clin. Pathol. 128(2), 245–254 (2007).
    • 45. Stower H. Tracing clear cell renal carcinoma evolution. Nat. Med. 24(6), 702 (2018).
    • 46. Almeida L, Silva R, Cavadas B et al. GLUT1, MCT1/4 and CD147 overexpression supports the metabolic reprogramming in papillary renal cell carcinoma. Histol Histopathol 32(10), 1029–1040 (2017).
    • 47. Amann T, Maegdefrau U, Hartmann A et al. GLUT1 expression is increased in hepatocellular carcinoma and promotes tumorigenesis. Am. J. Pathol. 174(4), 1544–1552 (2009).
    • 48. Sun HW, Yu XJ, Wu WC et al. GLUT1 and ASCT2 as predictors for prognosis of hepatocellular carcinoma. PLoS ONE 11(12), e0168907 (2016).
    • 49. Nishioka T, Oda Y, Seino Y et al. Distribution of the glucose transporters in human brain tumors. Cancer Res. 52(14), 3972–3979 (1992).
    • 50. Brown RS, Wahl RL. Overexpression of Glut-1 glucose transporter in human breast cancer. An immunohistochemical study. Cancer 72(10), 2979–2985 (1993).
    • 51. Shen YM, Arbman G, Olsson B, Sun XF. Overexpression of GLUT1 in colorectal cancer is independently associated with poor prognosis. Int. J. Biol. Markers 26(3), 166–172 (2011).
    • 52. Sasaki H, Shitara M, Yokota K et al. Overexpression of GLUT1 correlates with Kras mutations in lung carcinomas. Mol. Med. Rep. 5(3), 599–602 (2012).
    • 53. Reinicke K, Sotomayor P, Cisterna P, Delgado C, Nualart F, Godoy A. Cellular distribution of Glut-1 and Glut-5 in benign and malignant human prostate tissue. J. Cell Biochem. 113(2), 553–562 (2012).
    • 54. Kapoor K, Finer-Moore JS, Pedersen BP et al. Mechanism of inhibition of human glucose transporter GLUT1 is conserved between cytochalasin B and phenylalanine amides. Proc. Natl. Acad. Sci. USA 113(17), 4711–4716 (2016). •• This paper reports the crystal structure of cytochalasin B and GLUT1 protein complex and elucidates the binding site.
    • 55. Moreira L, Araujo I, Costa T et al. Quercetin and epigallocatechin gallate inhibit glucose uptake and metabolism by breast cancer cells by an estrogen receptor-independent mechanism. Exp. Cell. Res. 319(12), 1784–1795 (2013).
    • 56. Brito AF, Ribeiro M, Abrantes AM et al. New approach for treatment of primary liver tumors: the role of quercetin. Nutr. Cancer 68(2), 250–266 (2016).
    • 57. Azevedo C, Correia-Branco A, Araujo JR, Guimaraes JT, Keating E, Martel F. The chemopreventive effect of the dietary compound kaempferol on the MCF-7 human breast cancer cell line is dependent on inhibition of glucose cellular uptake. Nutr. Cancer 67(3), 504–513 (2015).
    • 58. Salas M, Obando P, Ojeda L et al. Resolution of the direct interaction with and inhibition of the human GLUT1 hexose transporter by resveratrol from its effect on glucose accumulation. Am. J. Physiol. Cell Physiol. 305(1), C90–99 (2013).
    • 59. Wu H, He L, Shi J et al. Resveratrol inhibits VEGF-induced angiogenesis in human endothelial cells associated with suppression of aerobic glycolysis via modulation of PKM2 nuclear translocation. Clin. Exp. Pharmacol. Physiol. 45(12), 1265–1273 (2018).
    • 60. Jung KH, Lee JH, Thien Quach CH et al. Resveratrol suppresses cancer cell glucose uptake by targeting reactive oxygen species-mediated hypoxia-inducible factor-1alpha activation. J. Nucl. Med. 54(12), 2161–2167 (2013).
    • 61. Gunnink LK, Alabi OD, Kuiper BD et al. Curcumin directly inhibits the transport activity of GLUT1. Biochimie 125, 179–185 (2016).
    • 62. Hu Y, Lou X, Wang R et al. Aspirin, a potential GLUT1 inhibitor in a vascular endothelial cell line. Open Med. 14, 552–560 (2019).
    • 63. Schimmer AD, Thomas MP, Hurren R et al. Identification of Small Molecules that Sensitize Resistant Tumor Cells to Tumor Necrosis Factor-Family Death Receptors. Cancer Res. 66(4), 2367–2375 (2006).
    • 64. Wood TE, Dalili S, Simpson CD et al. A novel inhibitor of glucose uptake sensitizes cells to FAS-induced cell death. Mol. Cancer Ther. 7(11), 3546–3555 (2008).
    • 65. Chan DA, Sutphin PD, Nguyen P et al. Targeting GLUT1 and the Warburg effect in renal cell carcinoma by chemical synthetic lethality. Sci. Transl. Med. 3(94), 94ra70 (2011).
    • 66. Kraus D, Reckenbeil J, Veit N et al. Targeting glucose transport and the NAD pathway in tumor cells with STF-31: a re-evaluation. Cell. Oncol. 41(5), 485–494 (2018).
    • 67. Liu Y, Cao Y, Zhang W et al. A small-molecule inhibitor of glucose transporter 1 downregulates glycolysis, induces cell-cycle arrest, and inhibits cancer cell growth in vitro and in vivo. Mol. Cancer Ther. 11(8), 1672–1682 (2012).
    • 68. Zhang W, Liu Y, Chen X, Bergmeier SC. Novel inhibitors of basal glucose transport as potential anticancer agents. Bioorg. Med. Chem. Lett. 20(7), 2191–2194 (2010).
    • 69. Ojelabi OA, Lloyd KP, Simon AH et al. WZB117 (2-fluoro-6-(m-hydroxybenzoyloxy) phenyl m-hydroxybenzoate) inhibits GLUT1-mediated sugar transport by binding reversibly at the exofacial sugar binding site. J. Biol. Chem. 291(52), 26762–26772 (2016).
    • 70. Siebeneicher H, Cleve A, Rehwinkel H et al. Identification and optimization of the first highly selective GLUT1 inhibitor BAY-876. ChemMedChem 11(20), 2261–2271 (2016). •• This paper describes the development process of the first highly selective GLUT1 inhibitor in detail.
    • 71. Siebeneicher H, Bauser M, Buchmann B et al. Identification of novel GLUT inhibitors. Bioorg. Med. Chem. Lett. 26(7), 1732–1737 (2016).
    • 72. Ung PM, Song W, Cheng L et al. Inhibitor discovery for the human GLUT1 from homology modeling and virtual screening. ACS Chem. Biol. 11(7), 1908–1916 (2016).
    • 73. Reckzeh ES, Karageorgis G, Schwalfenberg M et al. Inhibition of glucose transporters and glutaminase synergistically impairs tumor cell growth. Cell Chem. Biol. 26(9), 1214–1228 (2019).
    • 74. Kang SA, O'neill DJ, Machl AW et al. Discovery of small-molecule selective mTORC1 inhibitors via direct inhibition of glucose transporters. Cell Chem. Biol. 26(9), 1203–1213 (2019).
    • 75. Karageorgis G, Reckzeh ES, Ceballos J et al. Chromopynones are pseudo natural product glucose uptake inhibitors targeting glucose transporters GLUT-1 and -3. Nat. Chem. 10(11), 1103–1111 (2018).
    • 76. Ceballos J, Schwalfenberg M, Karageorgis G et al. Synthesis of indomorphan pseudo-natural product inhibitors of glucose transporters GLUT-1 and -3. Angew Chem. Int. Ed. Engl. 58(47), 17016–17025 (2019).
    • 77. Cuppoletti J, Mayhew E, Jung CY. Cytochalasin B binding to Ehrlich ascites tumor cells and its relationship to glucose carrier. Biochim. Biophys. Acta. 642(2), 392–404 (1981).
    • 78. Nelson KM, Dahlin JL, Bisson J et al. The essential medicinal chemistry of curcumin. J. Med. Chem. 60(5), 1620–1637 (2017).
    • 79. Unlu A, Nayir E, Dogukan Kalenderoglu M et al. Curcumin (turmeric) and cancer. J. Buon. 21(5), 1050–1060 (2016).
    • 80. Capodanno D, Angiolillo DJ. Aspirin for primary cardiovascular risk prevention and beyond in diabetes mellitus. Circulation 134(20), 1579–1594 (2016).
    • 81. Levine LD, Holland TL, Kim K et al. The role of aspirin and inflammation on reproduction: the EAGeR trial. Can. J. Physiol. Pharmacol. 97(3), 187–192 (2019).
    • 82. Liu YX, Feng JY, Sun MM et al. Aspirin inhibits the proliferation of hepatoma cells through controlling GLUT1-mediated glucose metabolism. Acta. Pharmacol. Sin. 40(1), 122–132 (2019).
    • 83. Wang T, Ning K, Lu TX, Hua D. Elevated expression of TrpC5 and GLUT1 is associated with chemoresistance in colorectal cancer. Oncol. Rep. 37(2), 1059–1065 (2017). •• This paper describes in detail the drug combination of wzb117 and 5-fluorouracil as GLUT1 inhibitor.
    • 84. Sawayama H, Ogata Y, Ishimoto T et al. Glucose transporter 1 regulates the proliferation and cisplatin sensitivity of esophageal cancer. Cancer Sci. 110(5), 1705–1714 (2019).