Skip to main content

EDITORIAL article

Front. Pharmacol., 30 July 2024
Sec. Experimental Pharmacology and Drug Discovery
This article is part of the Research Topic The Culprit Behind Some Diseases: Overexpression/hyperactivity of G6PD View all 5 articles

Editorial: The culprit behind some diseases: overexpression/hyperactivity of G6PD

  • 1Department of Molecular Biology and Genetics, Kilis 7 Aralik University, Kilis, Türkiye
  • 2School of Medicine, Department of Medical Biochemistry, Koc University, Istanbul, Türkiye
  • 3Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye

The pentose phosphate pathway (PPP) serves as a pivotal junction in cellular metabolism, playing a crucial role in regulating essential processes such as biosynthesis and oxidative defense. The primary factor for regulation in the activity of this pathway is glucose-6-phosphate dehydrogenase (G6PD), which is the first checkpoint enzyme of the pathway. Both hyperactivation and inactivation of this enzyme leads to metabolic imbalances. Alterations in the activity of the PPP related to hyper-activation and inactivation of the enzyme result in the occurrence of various diseases.

In this Research Topic, we aimed to focus on the critical role of the enzyme in metabolic diseases and to explore recent developments in this area. For this, we invited scientists studying diseases related to the G6PD enzyme. This collection includes important reviews, opinions, and original research articles.

Aging negatively affects mitochondrial activity, resulting in lower ATP generation and higher ROS levels. G6PD activity declines as the cell ages (Hodgkins, 2020), and the catalytic mechanism of the enzyme also changes during this period (Ulusu and Tandogan, 2006). This change causes oxidative stress in senescent cells. In this Research Topic, Yan et al. pointed out that aging-related changes in G6PD enzyme activity may result in neurodegenerative disorders. They also underline that the changing of the G6PD enzyme, which is a neuroprotective factor against the deleterious effect of endogenous ROS in the senescent period human brain, has an impact on the antioxidant system in Alzheimer’s disease. This is consistent with a previous study reported by Ulusu (2015).

G6PD is a housekeeping enzyme responsible for cell growth. During embryonic and organismal development, aberrant activation of the PPP or G6PD was observed (Wu et al., 2018b). Unlike these processes, hyperactive G6PD enzyme activity in a cell is related to tumor development (Wu et al., 2018a). There may be an upregulation of the expression of the G6PD enzyme due to many different factors such as mutation, pathogens, diseases etc. Whatever the reason, this situation can contribute to the formation or development of a tumor. Though the relationship between G6PD deficiency and infection has not been clearly understood until recent years, Sun et al. (2022) shed light on the dark side of the G6PD overexpression-infection relationship, which is one of the reasons for tumor formation/development, with a case report and literature review. Consequently, immunodeficiency resulting from G6PD deficiency causes infection. On the other side, the hyperactivity of the G6PD enzyme can be induced by both hereditary factors as well as infections. G6PD mRNA level and G6PD expression non-tumor liver tissue and tumor liver tissue infected by the Hepatitis B virus are stimulated according to non-infected liver tissues. Nonetheless, it has been demonstrated that G6PD expression level in tumor tissue is higher than in non-tumor tissue (Liu et al., 2015). In this Research Topic, Chang et al. have noted that G6PD enzyme hyperactivity is associated with another virus infection. According to their findings, human papillomavirus (HPV) causes overexpression of G6PD mRNA and G6PD protein associated with the HPV16 E6 protein in patients with cervical cancer. The same results were obtained in pcDNA-HPV16 E6 plasmid-transfected Hela cells. The relationship between infection and alterations in G6PD expression indicates that viral infections contribute to carcinogenesis by changing host G6PD expression.

Gene expression changes are among the most common reasons for G6PD overexpression. Overexpression of the G6PD enzyme may cause changes in the expression of many genes associated with different forms of cancer. In colon cancer, the G6PD enzyme is overexpressed due to upregulation of p21-activated kinase 4 (PAK4) (Zhang et al., 2017). PAK4, a member of the PAKs family, is frequently overexpressed in various tumor types and promotes carcinogenesis and progression (Wang et al., 2014) by enhancing cell growth and proliferation (Murray et al., 2010). This effect is caused by upregulating glucose metabolism as well as many other pathways. As a result, consumption of glucose and NADPH synthesis result in an increase in cancer cells. This situation is caused by increased G6PD enzyme activity as a result of PAK4’s mechanism of stopping G6PD inactivation via P53 by increasing P53 degradation of Mdm2 (Zhang et al., 2017). Another factor for G6PD overexpression is nuclear factor erythroid 2-related factor 2 (NRF2). However, its effect on G6PD expression is not straightforward but occurs through the regulation of transcription factor TAp73. This regulation occurs when NRF2 suppresses the E3 ligase PIRH2, hence prolonging the half-life of TAp73 (Zhang et al., 2022). TAp73 with enhanced half-life increases the expression of the G6PD enzyme (Du et al., 2013). Overexpression of the G6PD enzyme in cancer cells not only contributes to tumor development and formation but also causes the development of resistance to many treatment methods based on the principle of creating oxidative stress, such as chemotherapy and radiotherapy. Thus, the inhibition of enzyme activity is crucial for eliminating cancer produced by this hyperactivity. The G6PD enzyme has been a Research Topic of intense interest in the last decade due to its potential as a target of chemotherapeutic and chemo-sensitizer drug candidates for cancer treatment. Many compounds have been developed to decrease the hyperactivity of the enzyme in cancer cells, and their kinetic effects have been studied to inhibit the enzyme’s catalytic activity or prevent dimerization. The increase in the number of research articles published in the 2020s shows that the G6PD enzyme will be a common target in cancer treatment research. In this Research Topic, Xu et al. expressed that caffeine inhibits the G6PD enzyme by binding its structural NADP+ binding site. Caffeine reduces the formation of the dimer structure necessary for its catalytic activity. G6PD dimerization is potently related to the interaction between specific amino acids in the dimer interface. Critical residues such as T406, K403, and Y401 play a pivotal role in this process, and mutations in these amino acids can disrupt dimerization (Matte et al., 2020). In addition, the presence of NADP+ in the G6PD enzyme structure is another factor contributing to the dimerization process (Au et al., 1999).

Because the G6PD enzyme functions in the cell’s most crucial pathways, such as glycolysis, gluconeogenesis, PPP, and lipid metabolism, managing its activity is critical for an organism’s hemostasis. Expression of the G6PD enzyme varies depending on several regulators; it is also regulated by the control of hormones including insulin, glucocorticoids, and epidermal growth factor (EGF) in various tissues (Kletzien et al., 1994). Keran and Barker (1976) discovered that G6PD levels increase in the uterus of mature rats after 18 h following 5 µg/rat of estradiol (Keran and Barker, 1976). mRNA expression of the G6PD enzyme involved in carbohydrate and lipid metabolism downregulates with specific hormones (Pes and Dore, 2022). In the research performed on hormone-sensitive cell lines, androgen receptor (AR) was knocked down by using siRNAs in these cells. Following siRNA treatment, G6PD protein expression decreased. It has been determined that this effect is related to the motor regulation mediated by AR (Tsouko et al., 2014). A disruption in the endocrine system directly results in a change in the regulation of metabolic pathways, including PPP. Many chemicals impair endocrine function by binding hormone receptors (Schug et al., 2011; Lauretta et al., 2019), This change in the endocrine system causes a detrimental impact on human health (Diamanti-Kandarakis et al., 2009). This effect can be observed more effectively in energy metabolism and biosynthesis due to the roles of hormones in lipid, carbohydrate, and protein metabolism. There is limited research on the regulation of the G6PD enzyme by the endocrine system. In this Research Topic, Aydemir and Ulusu (2023) pointed out the endocrine-related regulation of G6PD expression. They expressed that endocrine-disrupting chemicals sourced from cosmetics, hygiene products, food and beverage packages, toys, and medical devices have impaired G6PD enzyme expression by effecting hormone metabolism. Individuals, particularly those who have G6PD deficiency, will experience metabolic and cardiovascular disorders as a result of the detrimental effects of EDC exposure throughout daily life on the G6PD enzyme.

In conclusion, this Research Topic provided detailed insights into the regulation of G6PD enzyme activity or expression, focusing on diseases related to G6PD enzyme hyperactivation. The overexpression in various disease states was discussed, with underlying causes and associated therapeutic strategies offering novel insights into treatment.

Author contributions

MK: Writing–original draft, Writing–review and editing. NU: Writing–review and editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Au, S. W., Naylor, C. E., Gover, S., Vandeputte-Rutten, L., Scopes, D. A., Mason, P. J., et al. (1999). Solution of the structure of tetrameric human glucose 6-phosphate dehydrogenase by molecular replacement. Acta Crystallogr. D. Biol. Crystallogr. 55, 826–834. doi:10.1107/s0907444999000827

PubMed Abstract | CrossRef Full Text | Google Scholar

Aydemir, D., and Ulusu, N. N. (2023). The impact of the endocrine-disrupting chemicals on the glucose-6-phosphate dehydrogenase enzyme activity. Front. Pharmacol. 14, 1133741. doi:10.3389/fphar.2023.1133741

PubMed Abstract | CrossRef Full Text | Google Scholar

Diamanti-Kandarakis, E., Bourguignon, J. P., Giudice, L. C., Hauser, R., Prins, G. S., Soto, A. M., et al. (2009). Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocr. Rev. 30, 293–342. doi:10.1210/er.2009-0002

PubMed Abstract | CrossRef Full Text | Google Scholar

Du, W., Jiang, P., Mancuso, A., Stonestrom, A., Brewer, M. D., Minn, A. J., et al. (2013). TAp73 enhances the pentose phosphate pathway and supports cell proliferation. Nat. Cell Biol. 15, 991–1000. doi:10.1038/ncb2789

PubMed Abstract | CrossRef Full Text | Google Scholar

Hodgkins, S. R. (2020). “Intrinsic defects leading to increased erythrocyte destruction,” in Rodak's hematology: clinical principles and applications. Editor E. M. KEOHANE 6th ed. (Canada: Elsevier, Inc).

CrossRef Full Text | Google Scholar

Keran, E. E., and Barker, K. L. (1976). Regulation of glucose-6-phosphate dehydrogenase activity in uterine tissue in organ culture. Endocrinology 99, 1386–1397. doi:10.1210/endo-99-5-1386

PubMed Abstract | CrossRef Full Text | Google Scholar

Kletzien, R. F., Harris, P. K., and Foellmi, L. A. (1994). Glucose-6-phosphate dehydrogenase: a "housekeeping" enzyme subject to tissue-specific regulation by hormones, nutrients, and oxidant stress. FASEB J. 8, 174–181. doi:10.1096/fasebj.8.2.8119488

PubMed Abstract | CrossRef Full Text | Google Scholar

Lauretta, R., Sansone, A., Sansone, M., Romanelli, F., and Appetecchia, M. (2019). Endocrine disrupting chemicals: effects on endocrine glands. Front. Endocrinol. (Lausanne) 10, 178. doi:10.3389/fendo.2019.00178

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, B., Fang, M., He, Z., Cui, D., Jia, S., Lin, X., et al. (2015). Hepatitis B virus stimulates G6PD expression through HBx-mediated Nrf2 activation. Cell Death Dis. 6, e1980. doi:10.1038/cddis.2015.322

PubMed Abstract | CrossRef Full Text | Google Scholar

Matte, A., Lupo, F., Tibaldi, E., Di Paolo, M. L., Federti, E., Carpentieri, A., et al. (2020). Fyn specifically Regulates the activity of red cell glucose-6-phosphate-dehydrogenase. Redox Biol. 36, 101639. doi:10.1016/j.redox.2020.101639

PubMed Abstract | CrossRef Full Text | Google Scholar

Murray, B. W., Guo, C., Piraino, J., Westwick, J. K., Zhang, C., Lamerdin, J., et al. (2010). Small-molecule p21-activated kinase inhibitor PF-3758309 is a potent inhibitor of oncogenic signaling and tumor growth. Proc. Natl. Acad. Sci. U. S. A. 107, 9446–9451. doi:10.1073/pnas.0911863107

PubMed Abstract | CrossRef Full Text | Google Scholar

Pes, G. M., and Dore, M. P. (2022). Acquired glucose-6-phosphate dehydrogenase deficiency. J. Clin. Med. 11, 6689. doi:10.3390/jcm11226689

PubMed Abstract | CrossRef Full Text | Google Scholar

Schug, T. T., Janesick, A., Blumberg, B., and Heindel, J. J. (2011). Endocrine disrupting chemicals and disease susceptibility. J. Steroid Biochem. Mol. Biol. 127, 204–215. doi:10.1016/j.jsbmb.2011.08.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, B., Li, Q., Dong, X., Hou, J., Wang, W., Ying, W., et al. (2022). Severe G6PD deficiency leads to recurrent infections and defects in ROS production: case report and literature review. Front. Genet. 13, 1035673. doi:10.3389/fgene.2022.1035673

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsouko, E., Khan, A. S., White, M. A., Han, J. J., Shi, Y., Merchant, F. A., et al. (2014). Regulation of the pentose phosphate pathway by an androgen receptor-mTOR-mediated mechanism and its role in prostate cancer cell growth. Oncogenesis 3, e103. doi:10.1038/oncsis.2014.18

PubMed Abstract | CrossRef Full Text | Google Scholar

Ulusu, N. N. (2015). Glucose-6-phosphate dehydrogenase deficiency and Alzheimer's disease: partners in crime? The hypothesis. Med. Hypotheses 85, 219–223. doi:10.1016/j.mehy.2015.05.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Ulusu, N. N., and Tandogan, B. (2006). Purification and kinetics of sheep kidney cortex glucose-6-phosphate dehydrogenase. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 143, 249–255. doi:10.1016/j.cbpb.2005.11.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C., Li, Y., Zhang, H., Liu, F., Cheng, Z., Wang, D., et al. (2014). Oncogenic PAK4 regulates Smad2/3 axis involving gastric tumorigenesis. Oncogene 33, 3473–3484. doi:10.1038/onc.2013.300

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y. H., Lee, Y. H., Shih, H. Y., Chen, S. H., Cheng, Y. C., and Tsun-Yee Chiu, D. (2018b). Glucose-6-phosphate dehydrogenase is indispensable in embryonic development by modulation of epithelial-mesenchymal transition via the NOX/Smad3/miR-200b axis. Cell Death Dis. 9, 10. doi:10.1038/s41419-017-0005-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, S., Wang, H., Li, Y., Xie, Y., Huang, C., Zhao, H., et al. (2018a). Transcription factor YY1 promotes cell proliferation by directly activating the pentose phosphate pathway. Cancer Res. 78, 4549–4562. doi:10.1158/0008-5472.CAN-17-4047

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Zhang, X., Li, Y., Shao, Y., Xiao, J., Zhu, G., et al. (2017). PAK4 regulates G6PD activity by p53 degradation involving colon cancer cell growth. Cell Death Dis. 8, e2820. doi:10.1038/cddis.2017.85

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Xu, Y., Lu, W., Li, J., Yu, S., Brown, E. J., et al. (2022). G6PD-mediated increase in de novo NADP(+) biosynthesis promotes antioxidant defense and tumor metastasis. Sci. Adv. 8, eabo0404. doi:10.1126/sciadv.abo0404

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: glucose 6 phosphate dehydrogenase, Oxitative stress, NADPH, overexpression of G6PD, alzhaimer disease, cancer, metabolic disease

Citation: Karaman M and Ulusu NN (2024) Editorial: The culprit behind some diseases: overexpression/hyperactivity of G6PD. Front. Pharmacol. 15:1459741. doi: 10.3389/fphar.2024.1459741

Received: 04 July 2024; Accepted: 12 July 2024;
Published: 30 July 2024.

Edited and reviewed by:

Filippo Drago, University of Catania, Italy

Copyright © 2024 Karaman and Ulusu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Muhammet Karaman, mhmmtkaraman@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.