The Function of GPT2 in Tumor Progression

Authors

  • Jun Xie Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
  • Hongjin Shi Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
  • Huake Wang Department of Urology, Kunming Children’s Hospital, Kunming, China
  • Chong Li Institute of Biophysics, Chinese Academy of Sciences, Beijing, China;Zhongke Jianlan Medical Research Institute, Beijing, China
  • Haifeng Wang Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China

DOI:

https://doi.org/10.18063/cp.393

Keywords:

Glutamate pyruvate transaminase 2, Glutamine, Isoenzymes

Abstract

Glutamate pyruvate transaminase 2 (GPT2) is one of the GPTs and is widely used as a biomarker of hepatocellular injury, along with GPT1. GPT2, a glutamine-metabolizing transaminase found in mitochondria, catalyzes the reversible process between glutamate, pyruvate, α-ketoglutarate, and alanine. Compared to GPT1, the intracellular abundance of GPT2 is higher, suggesting that its enzymatic activity has a considerable role in glucose metabolism, amino acid metabolism, and lipid metabolism. In recent years, it has been discovered that deletion or mutation of GPT2 causes malignant transformation of tumors and that its expression level is closely correlated with tumor development. It is for this reason that the level of GPT2 can be used to reflect the metabolism level of the tumor cells in the body and can indicate metastasis based on its changes. The metabolism level of GPT2 in tumor cells is expected to be a marker in the tumor diagnostic process and subsequently contribute to early detection, thus improving tumor diagnosis and patient prognosis. This paper presents an overview of the current state of GPT2 research in the progression of tumors.

 

References

Mirestean CC, Iancu RI, Iancu D, 2022, New Horizons in Modulating the Radio-sensitivity of Head and Neck Cancer-100 Years after Warburg’ Effect Discovery. Front Oncol, 12:908695. DOI: 10.3389/fonc.2022.908695

Kao TW, Chuang YC, Lee HL, et al., 2022, Therapeutic Targeting of Glutaminolysis as a Novel Strategy to Combat Cancer Stem Cells. Int J Mol Sci, 23:15296. DOI: 10.3390/ijms232315296

Niklison-Chirou MV, Erngren I, Engskog M, et al., 2017, TAp73 is a Marker of Glutamine Addiction in Medulloblastoma. Genes Dev, 31:1738–53. DOI: 10.1101/gad.302349.117

Yang L, Venneti S, Nagrath D, 2017, Glutaminolysis: A Hallmark of Cancer Metabolism. Annu Rev Biomed Eng, 19:163–94. DOI: 10.1146/annurev-bioeng-071516-044546

Li T, Le A, 2018, Glutamine Metabolism in Cancer. Adv Exp Med Biol, 1063:13–32. DOI: 10.1007/978-3-319-77736-8_2

Cicatiello AG, Sagliocchi S, Nappi A, et al., 2022, Thyroid Hormone Regulates Glutamine Metabolism and Anaplerotic Fluxes by Inducing Mitochondrial Glutamate Aminotransferase GPT2. Cell Rep, 38:110409. DOI: 10.1016/j.celrep.2022.110409

Kim M, Gwak J, Hwang S, et al., 2019, Mitochondrial GPT2 Plays a Pivotal Role in Metabolic Adaptation to the Perturbation of Mitochondrial Glutamine Metabolism. Oncogene, 38:4729–38. DOI: 10.1038/s41388-019-0751-4

Sajnani K, Islam F, Smith RA, et al., 2017, Genetic Alterations in Krebs Cycle and Its Impact on Cancer Pathogenesis. Biochimie, 135:164–72. DOI: 10.1016/j.biochi.2017.02.008

Luengo A, Gui DY, Heiden MG, 2017, Targeting Metabolism for Cancer Therapy. Cell Chem Biol, 24:1161–80. DOI: 10.1016/j.chembiol.2017.08.028

Zhao H, Wu W, Li X, et al., 2022, Long Noncoding RNA UCA1 Promotes Glutamine-driven Anaplerosis of Bladder Cancer by Interacting with hnRNP I/L to Upregulate GPT2 Expression. Transl Oncol, 17:101340. DOI: 10.1016/j.tranon.2022.10134

Gondas E, Hives M, Kliment J, et al., 2022, The Ubiquitous Expression of Pyruvate Carboxylase among Human Prostate Tumors. Bratisl Lek Listy, 123:487–90. DOI: 10.4149/BLL_2022_077

Bernfeld E, Foster DA, 2019, Glutamine as an Essential Amino Acid for KRas-Driven Cancer Cells. Trends Endocrinol Metab, 30:357–68. DOI: 10.1016/j.tem.2019.03.003

Bodineau C, Tomé M, Murdoch PD, et al., 2022, Glutamine, MTOR and Autophagy: A Multiconnection Relationship. Autophagy, 18:2749–50. DOI: 10.1080/15548627.2022.2062875

Yang X, Li Z, Ren H, et al., 2022, New Progress of Glutamine Metabolism in the Occurrence, Development, and Treatment of Ovarian Cancer from Mechanism to Clinic. Front Oncol, 12:1018642. DOI: 10.3389/fonc.2022.1018642

Kodama M, Oshikawa K, Shimizu H, et al., 2020, A Shift in Glutamine Nitrogen Metabolism Contributes to the Malignant Progression of Cancer. Nat Commun, 11:1320. DOI: 10.1038/s41467-020-15136-9

Hakvoort TB, He Y, Kulik W, et al., 2017, Pivotal Role of Glutamine Synthetase in Ammonia Detoxification. Hepatology, 65:281–93. DOI: 10.1002/hep.28852

Soria LR, Nitzahn M, De Angelis A, et al., 2019, Hepatic Glutamine Synthetase Augmentation Enhances Ammonia Detoxification. J Inherit Metab Dis, 42:1128–35. DOI: 10.1002/jimd.12070

Mukherjee A, Ahmed N, Rose FT, et al., 2020, Asparagine Synthetase is Highly Expressed at Baseline in the Pancreas through Heightened PERK Signaling. Cell Mol Gastroenterol Hepatol, 9:1–13. DOI: 10.1016/j.jcmgh.2019.08.003

Fan S, Wang Y, Zhang Z, et al., 2018, High Expression of Glutamate-ammonia Ligase is Associated with Unfavorable Prognosis in Patients with Ovarian Cancer. J Cell Biochem, 119:6008–15. DOI: 10.1002/jcb.26797

Moudi B, Heidari Z, Mahmoudzadeh-Sagheb H, et al., 2018, Concomitant Use of Heat-shock Protein 70, Glutamine Synthetase and Glypican-3 is Useful in Diagnosis of HBV-related Hepatocellular Carcinoma with Higher Specificity and Sensitivity. Eur J Histochem, 62:2859. DOI: 10.4081/ejh.2018.2859

Ye J, Huang Q, Xu J, et al., 2018, Targeting of Glutamine Transporter ASCT2 and Glutamine Synthetase Suppresses Gastric Cancer Cell Growth. J Cancer Res Clin Oncol, 144: 821–33. DOI: 10.1007/s00432-018-2605-9

Eelen G, Dubois C, Cantelmo AR, et al., 2018, Role of Glutamine Synthetase in Angiogenesis Beyond Glutamine Synthesis. Nature, 561:63–9. DOI: 10.1038/s41586-018-0466-7

Palmieri EM, Menga A, Martín-Pérez R, et al., 2017, Pharmacologic or Genetic Targeting of Glutamine Synthetase Skews Macrophages toward an M1-like Phenotype and Inhibits Tumor Metastasis. Cell Rep, 20:1654–66. DOI: 10.1016/j.celrep.2017.07.054

Matés JM, Di Paola FJ, Campos-Sandoval JA, et al., 2020, Therapeutic Targeting of Glutaminolysis as an Essential Strategy to Combat Cancer. Semin Cell Dev Biol, 98:34–43. DOI: 10.1016/j.semcdb.2019.05.012

Katt WP, Lukey MJ, Cerione RA, 2017, A Tale of Two Glutaminases: Homologous Enzymes with Distinct Roles in Tumorigenesis. Future Med Chem, 9:223–43. DOI: 10.4155/fmc-2016-0190

Cluntun AA, Lukey MJ, Cerione RA, et al., 2017, Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer, 3:169–80. DOI: 10.1016/j.trecan.2017.01.005

Sengupta D, Cassel T, Teng KY, et al., 2020, Regulation of Hepatic Glutamine Metabolism by miR-122. Mol Metab, 34:174–86. DOI: 10.1016/j.molmet.2020.01.003

Zhang J, Wang L, Mao S, et al., 2018, miR-1-3p Contributes to Cell Proliferation and Invasion by Targeting Glutaminase in Bladder Cancer Cells. Cell Physiol Biochem, 51:513–27. DOI: 10.1159/00049527

Cao Y, Lin SH, Wang Y, et al., 2017, Glutamic Pyruvate Transaminase GPT2 Promotes Tumorigenesis of Breast Cancer Cells by Activating Sonic Hedgehog Signaling. Theranostics, 7:3021–33. DOI: 10.7150/thno.18992

Mitra D, Vega-Rubin-de-Celis S, Royla N, et al., 2021, Abrogating GPT2 in Triple-negative Breast Cancer Inhibits Tumor Growth and Promotes Autophagy. Int J Cancer, 148:1993–2009. DOI: 10.1002/ijc.33456

Xu P, Oosterveer MH, Stein S, et al., 2016, LRH-1- dependent Programming of Mitochondrial Glutamine Processing Drives Liver Cancer. Genes Dev, 30:1255–60. DOI: 10.1101/gad.277483.11

Wang R, Xiang W, Xu Y, et al., 2020, Enhanced Glutamine Utilization Mediated by SLC1A5 and GPT2 is an Essential Metabolic Feature of Colorectal Signet Ring Cell Carcinoma with Therapeutic Potential. Ann Transl Med, 8:302. DOI: 10.21037/atm.2020.03.31

Chen W, Dai G, Qian Y, et al., 2022, PIK3CA Mutation Affects the Proliferation of Colorectal Cancer Cells through the PI3K-MEK/PDK1-GPT2 Pathway. Oncol Rep, 47:11. DOI: 10.3892/or.2021.8222

Caiola E, Colombo M, Sestito G, et al., 2020, Glutaminase Inhibition on NSCLC Depends on Extracellular Alanine Exploitation. Cells, 9:1766. DOI: 10.3390/cells9081766

Wu J, Miao C, Wang Y, et al., 2022, SPTBN1 Abrogates Renal Clear Cell Carcinoma Progression Via Glycolysis Reprogramming in a GPT2-Dependent Manner. J Transl Med, 20:603. DOI: 10.1186/or.2022.12.16

Franco YE, Alves MJ, et al., 2021, Glutaminolysis Dynamics During Astrocytoma Progression Correlates with Tumor Aggressiveness. Cancer Metab, 9:18. DOI: 10.1186/or.2021.04.28

Downloads

Published

2023-03-03