Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

GPER-mediated stabilization of HIF-1α contributes to upregulated aerobic glycolysis in tamoxifen-resistant cells

Abstract

Tamoxifen is a first-line therapeutic drug for oestrogen-receptor positive breast cancer; however, like other therapeutics, its clinical use is limited by acquired resistance. Tamoxifen-resistant cells have demonstrated enhanced aerobic glycolysis; however, the mechanisms underlying this upregulation remain unclear. Here, we demonstrated that G-protein coupled oestrogen receptor (GPER) was involved in the upregulation of aerobic glycolysis via induction of hypoxia-inducible factor-1α (HIF-1α) expression and transcriptional activity in tamoxifen-resistant cells. Additionally, GPER stabilized HIF-1α through inhibiting its hydroxylation and ubiquitin-mediated degradation, which were associated with upregulation of C-terminal hydrolase-L1 (UCH-L1), downregulation of prolyl hydroxylase 2 (PHD2) and von Hippel-Lindau tumour suppressor protein (pVHL), induction of HIF-1α/UCH-L1 interaction, and suppression of HIF-1α/PHD2-pVHL association. The GPER/HIF-1α axis was functionally responsible for regulating tamoxifen sensitivity both in vitro and in vivo. Moreover, there was a positive correlation between GPER and HIF-1α expression in clinical breast cancer tissues, and high levels of GPER combined with nuclear HIF-1α indicated poor overall survival. High levels of the GPER/HIF-1α axis were also correlated with shorter relapse-free survival in patients receiving tamoxifen. Hence, our findings support a critical role of GPER/HIF-1α axis in the regulation of aerobic glycolysis in tamoxifen-resistant cells, offering a potential therapeutic target for tamoxifen-resistant breast cancer.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: GPER is required for glycolysis in tamoxifen-resistant breast cancer cells.
Fig. 2: GPER mediates glycolysis via upregulation of HIF-1α expression and transcriptional activity.
Fig. 3: GPER increases HIF-1α protein stability by suppressing ubiquitin degradation.
Fig. 4: GPER enhances HIF-1α/UCH-L1 interaction and suppressed HIF-1α/PHD2-pVHL interaction.
Fig. 5: GPER/HIF-1α axis is involved in regulating tamoxifen resistance.
Fig. 6: GPER/HIF-1α axis is involved in tamoxifen resistance in vivo.
Fig. 7: Positive correlation between GPER and HIF-1α.
Fig. 8: Schematic diagram of GPER/HIF-1α axis in upregulated aerobic glycolysis in tamoxifen-resistant cells.

Similar content being viewed by others

Data availability

The datasets generated and analysed during the current study are available from the corresponding author on reasonable request.

References

  1. Barua D, Gupta A, Gupta S. Targeting the IRE1-XBP1 axis to overcome endocrine resistance in breast cancer: Opportunities and challenges. Cancer Lett. 2020;486:29–37.

    Article  CAS  Google Scholar 

  2. Najim O, Seghers S, Sergoynne L, Van Gaver H, Papadimitriou K, Wouters K, et al. The association between type of endocrine therapy and development of estrogen receptor-1 mutation(s) in patients with hormone-sensitive advanced breast cancer: A systematic review and meta-analysis of randomized and non-randomized trials. Biochim Biophys Acta Rev Cancer. 2019;1872:188315.

    Article  CAS  Google Scholar 

  3. Powles TJ, Ashley S, Tidy A, Smith IE, Dowsett M. Twenty-year follow-up of the Royal Marsden randomized, double-blinded tamoxifen breast cancer prevention trial. J Natl Cancer Inst. 2007;99:283–90.

    Article  CAS  Google Scholar 

  4. Chang M. Tamoxifen resistance in breast cancer. Biomol Ther (Seoul). 2012;20:256–67.

    Article  CAS  Google Scholar 

  5. Liu SS, Li Y, Zhang H, Zhang D, Zhang XB, Wang X, et al. The ERalpha-miR-575-p27 feedback loop regulates tamoxifen sensitivity in ER-positive Breast Cancer. Theranostics. 2020;10:10729–42.

    Article  CAS  Google Scholar 

  6. Yao J, Deng K, Huang J, Zeng R, Zuo J. Progress in the Understanding of the Mechanism of Tamoxifen Resistance in Breast Cancer. Front Pharm. 2020;11:592912.

    Article  CAS  Google Scholar 

  7. Ranganathan P, Nadig N, Nambiar S. Non-canonical Estrogen Signaling in Endocrine Resistance. Front Endocrinol (Lausanne). 2019;10:708.

    Article  Google Scholar 

  8. Vallee A, Lecarpentier Y, Vallee JN. The Key Role of the WNT/beta-Catenin Pathway in Metabolic Reprogramming in Cancers under Normoxic Conditions. Cancers (Basel). 2021;13:5557.

  9. Vaupel P, Multhoff G. Revisiting the Warburg effect: historical dogma versus current understanding. J Physiol. 2021;599:1745–57.

    Article  CAS  Google Scholar 

  10. Abdel-Wahab AF, Mahmoud W, Al-Harizy RM. Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharm Res. 2019;150:104511.

    Article  CAS  Google Scholar 

  11. San-Millan I, Brooks GA. Reexamining cancer metabolism: lactate production for carcinogenesis could be the purpose and explanation of the Warburg Effect. Carcinogenesis. 2017;38:119–33.

    CAS  Google Scholar 

  12. Ma L, Zong X. Metabolic Symbiosis in Chemoresistance: Refocusing the Role of Aerobic Glycolysis. Front Oncol. 2020;10:5.

    Article  Google Scholar 

  13. He M, Jin Q, Chen C, Liu Y, Ye X, Jiang Y, et al. The miR-186-3p/EREG axis orchestrates tamoxifen resistance and aerobic glycolysis in breast cancer cells. Oncogene. 2019;38:5551–65.

    Article  CAS  Google Scholar 

  14. Das CK, Parekh A, Parida PK, Bhutia SK, Mandal M. Lactate dehydrogenase A regulates autophagy and tamoxifen resistance in breast cancer. Biochim Biophys Acta Mol Cell Res. 2019;1866:1004–18.

    Article  CAS  Google Scholar 

  15. Daurio NA, Tuttle SW, Worth AJ, Song EY, Davis JM, Snyder NW, et al. AMPK Activation and Metabolic Reprogramming by Tamoxifen through Estrogen Receptor-Independent Mechanisms Suggests New Uses for This Therapeutic Modality in Cancer Treatment. Cancer Res. 2016;76:3295–306.

    Article  CAS  Google Scholar 

  16. Padro M, Louie RJ, Lananna BV, Krieg AJ, Timmerman LA, Chan DA. Genome-independent hypoxic repression of estrogen receptor alpha in breast cancer cells. BMC Cancer. 2017;17:203.

    Article  Google Scholar 

  17. Kierans SJ, Taylor CT. Regulation of glycolysis by the hypoxia-inducible factor (HIF): implications for cellular physiology. J Physiol. 2021;599:23–37.

    Article  CAS  Google Scholar 

  18. Gruber G, Greiner RH, Hlushchuk R, Aebersold DM, Altermatt HJ, Berclaz G, et al. Hypoxia-inducible factor 1 alpha in high-risk breast cancer: an independent prognostic parameter? Breast Cancer Res 2004;6:R191–8.

    Article  CAS  Google Scholar 

  19. Generali D, Buffa FM, Berruti A, Brizzi MP, Campo L, Bonardi S, et al. Phosphorylated ERalpha, HIF-1alpha, and MAPK signaling as predictors of primary endocrine treatment response and resistance in patients with breast cancer. J Clin Oncol. 2009;27:227–34.

    Article  Google Scholar 

  20. Jogi A, Ehinger A, Hartman L, Alkner S. Expression of HIF-1alpha is related to a poor prognosis and tamoxifen resistance in contralateral breast cancer. PLoS One. 2019;14:e0226150.

    Article  Google Scholar 

  21. Yang J, AlTahan A, Jones DT, Buffa FM, Bridges E, Interiano RB, et al. Estrogen receptor-alpha directly regulates the hypoxia-inducible factor 1 pathway associated with antiestrogen response in breast cancer. Proc Natl Acad Sci USA. 2015;112:15172–7.

    Article  CAS  Google Scholar 

  22. Kim MR, Choi HS, Yang JW, Park BC, Kim JA, Kang KW. Enhancement of vascular endothelial growth factor-mediated angiogenesis in tamoxifen-resistant breast cancer cells: role of Pin1 overexpression. Mol Cancer Ther. 2009;8:2163–71.

    Article  CAS  Google Scholar 

  23. Woo YM, Shin Y, Lee EJ, Lee S, Jeong SH, Kong HK, et al. Inhibition of Aerobic Glycolysis Represses Akt/mTOR/HIF-1alpha Axis and Restores Tamoxifen Sensitivity in Antiestrogen-Resistant Breast Cancer Cells. PLoS One. 2015;10:e0132285.

    Article  Google Scholar 

  24. Cowman SJ, Koh MY. Revisiting the HIF switch in the tumor and its immune microenvironment. Trends Cancer. 2022;8:28–42.

    Article  CAS  Google Scholar 

  25. Hsu LH, Chu NM, Lin YF, Kao SH. G-Protein Coupled Estrogen Receptor in Breast Cancer. Int J Mol Sci. 2019;20:306.

  26. Filardo EJ, Graeber CT, Quinn JA, Resnick MB, Giri D, DeLellis RA, et al. Distribution of GPR30, a seven membrane-spanning estrogen receptor, in primary breast cancer and its association with clinicopathologic determinants of tumor progression. Clin Cancer Res. 2006;12:6359–66.

    Article  CAS  Google Scholar 

  27. Jacenik D, Cygankiewicz AI, Krajewska WM. The G protein-coupled estrogen receptor as a modulator of neoplastic transformation. Mol Cell Endocrinol. 2016;429:10–8.

    Article  CAS  Google Scholar 

  28. Ignatov T, Treeck O, Kalinski T, Ortmann O, Ignatov A. GPER-1 expression is associated with a decreased response rate to primary tamoxifen therapy of breast cancer patients. Arch Gynecol Obstet. 2020;301:565–71.

    Article  CAS  Google Scholar 

  29. Ignatov T, Claus M, Nass N, Haybaeck J, Seifert B, Kalinski T, et al. G-protein-coupled estrogen receptor GPER-1 expression in hormone receptor-positive breast cancer is associated with poor benefit of tamoxifen. Breast Cancer Res Treat. 2019;174:121–7.

    Article  CAS  Google Scholar 

  30. Mo Z, Liu M, Yang F, Luo H, Li Z, Tu G, et al. GPR30 as an initiator of tamoxifen resistance in hormone-dependent breast cancer. Breast Cancer Res. 2013;15:R114.

    Article  Google Scholar 

  31. Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science. 2001;292:468–72.

    Article  CAS  Google Scholar 

  32. Masson N, Willam C, Maxwell PH, Pugh CW, Ratcliffe PJ. Independent function of two destruction domains in hypoxia-inducible factor-alpha chains activated by prolyl hydroxylation. EMBO J. 2001;20:5197–206.

    Article  CAS  Google Scholar 

  33. Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, et al. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol. 2000;2:423–7.

    Article  CAS  Google Scholar 

  34. Goto Y, Zeng L, Yeom CJ, Zhu Y, Morinibu A, Shinomiya K, et al. UCHL1 provides diagnostic and antimetastatic strategies due to its deubiquitinating effect on HIF-1alpha. Nat Commun. 2015;6:6153.

    Article  CAS  Google Scholar 

  35. Wang LJ, Han SX, Bai E, Zhou X, Li M, Jing GH, et al. Dose-dependent effect of tamoxifen in tamoxifen-resistant breast cancer cells via stimulation by the ERK1/2 and AKT signaling pathways. Oncol Rep. 2013;29:1563–9.

    Article  CAS  Google Scholar 

  36. Yuan J, Liu M, Yang L, Tu G, Zhu Q, Chen M, et al. Acquisition of epithelial-mesenchymal transition phenotype in the tamoxifen-resistant breast cancer cell: a new role for G protein-coupled estrogen receptor in mediating tamoxifen resistance through cancer-associated fibroblast-derived fibronectin and beta1-integrin signaling pathway in tumor cells. Breast Cancer Res. 2015;17:69.

    Article  Google Scholar 

  37. Ignatov A, Ignatov T, Roessner A, Costa SD, Kalinski T. Role of GPR30 in the mechanisms of tamoxifen resistance in breast cancer MCF-7 cells. Breast Cancer Res Treat. 2010;123:87–96.

    Article  CAS  Google Scholar 

  38. Wang M, Chen W, Chen J, Yuan S, Hu J, Han B, et al. Abnormal saccharides affecting cancer multi-drug resistance (MDR) and the reversal strategies. Eur J Med Chem. 2021;220:113487.

    Article  CAS  Google Scholar 

  39. Fischer K, Hoffmann P, Voelkl S, Meidenbauer N, Ammer J, Edinger M, et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood. 2007;109:3812–9.

    Article  CAS  Google Scholar 

  40. Martinez-Outschoorn UE, Prisco M, Ertel A, Tsirigos A, Lin Z, Pavlides S, et al. Ketones and lactate increase cancer cell “stemness,” driving recurrence, metastasis and poor clinical outcome in breast cancer: achieving personalized medicine via Metabolo-Genomics. Cell Cycle. 2011;10:1271–86.

    Article  CAS  Google Scholar 

  41. Liu X, Miao W, Huang M, Li L, Dai X, Wang Y. Elevated Hexokinase II Expression Confers Acquired Resistance to 4-Hydroxytamoxifen in Breast Cancer Cells. Mol Cell Proteom. 2019;18:2273–84.

    Article  CAS  Google Scholar 

  42. Raez LE, Papadopoulos K, Ricart AD, Chiorean EG, Dipaola RS, Stein MN, et al. A phase I dose-escalation trial of 2-deoxy-D-glucose alone or combined with docetaxel in patients with advanced solid tumors. Cancer Chemother Pharm. 2013;71:523–30.

    Article  CAS  Google Scholar 

  43. Molina L, Bustamante F, Ortloff A, Ramos I, Ehrenfeld P, Figueroa CD. Continuous Exposure of Breast Cancer Cells to Tamoxifen Upregulates GPER-1 and Increases Cell Proliferation. Front Endocrinol (Lausanne). 2020;11:563165.

    Article  Google Scholar 

  44. Catalano S, Giordano C, Panza S, Chemi F, Bonofiglio D, Lanzino M, et al. Tamoxifen through GPER upregulates aromatase expression: a novel mechanism sustaining tamoxifen-resistant breast cancer cell growth. Breast Cancer Res Treat. 2014;146:273–85.

    Article  CAS  Google Scholar 

  45. Ignatov A, Ignatov T, Weissenborn C, Eggemann H, Bischoff J, Semczuk A, et al. G-protein-coupled estrogen receptor GPR30 and tamoxifen resistance in breast cancer. Breast Cancer Res Treat. 2011;128:457–66.

    Article  CAS  Google Scholar 

  46. Yu T, Cheng H, Ding Z, Wang Z, Zhou L, Zhao P, et al. GPER mediates decreased chemosensitivity via regulation of ABCG2 expression and localization in tamoxifen-resistant breast cancer cells. Mol Cell Endocrinol. 2020;506:110762.

    Article  CAS  Google Scholar 

  47. Sjostrom M, Hartman L, Grabau D, Fornander T, Malmstrom P, Nordenskjold B, et al. Lack of G protein-coupled estrogen receptor (GPER) in the plasma membrane is associated with excellent long-term prognosis in breast cancer. Breast Cancer Res Treat. 2014;145:61–71.

    Article  Google Scholar 

  48. Johnston SR, Saccani-Jotti G, Smith IE, Salter J, Newby J, Coppen M, et al. Changes in estrogen receptor, progesterone receptor, and pS2 expression in tamoxifen-resistant human breast cancer. Cancer Res. 1995;55:3331–8.

    CAS  Google Scholar 

  49. Yin H, Zhu Q, Liu M, Tu G, Li Q, Yuan J, et al. GPER promotes tamoxifen-resistance in ER+ breast cancer cells by reduced Bim proteins through MAPK/Erk-TRIM2 signaling axis. Int J Oncol. 2017;51:1191–8.

    Article  CAS  Google Scholar 

  50. Yu T, Yang G, Hou Y, Tang X, Wu C, Wu XA, et al. Cytoplasmic GPER translocation in cancer-associated fibroblasts mediates cAMP/PKA/CREB/glycolytic axis to confer tumor cells with multidrug resistance. Oncogene. 2017;36:2131–45.

    Article  CAS  Google Scholar 

  51. Alam MW, Persson CU, Reinbothe S, Kazi JU, Ronnstrand L, Wigerup C, et al. HIF2alpha contributes to antiestrogen resistance via positive bilateral crosstalk with EGFR in breast cancer cells. Oncotarget. 2016;7:11238–50.

    Article  Google Scholar 

  52. Hoefflin R, Harlander S, Schafer S, Metzger P, Kuo F, Schonenberger D, et al. HIF-1alpha and HIF-2alpha differently regulate tumour development and inflammation of clear cell renal cell carcinoma in mice. Nat Commun. 2020;11:4111.

    Article  CAS  Google Scholar 

  53. Wang V, Davis DA, Haque M, Huang LE, Yarchoan R. Differential gene up-regulation by hypoxia-inducible factor-1alpha and hypoxia-inducible factor-2alpha in HEK293T cells. Cancer Res. 2005;65:3299–306.

    Article  CAS  Google Scholar 

  54. De Francesco EM, Pellegrino M, Santolla MF, Lappano R, Ricchio E, Abonante S, et al. GPER mediates activation of HIF1alpha/VEGF signaling by estrogens. Cancer Res. 2014;74:4053–64.

    Article  Google Scholar 

  55. Zhang L, Xiong W, Li N, Liu H, He H, Du Y, et al. Estrogen stabilizes hypoxia-inducible factor 1alpha through G protein-coupled estrogen receptor 1 in eutopic endometrium of endometriosis. Fertil Steril. 2017;107:439–47.

    Article  CAS  Google Scholar 

  56. De Francesco EM, Maggiolini M, Musti AM. Crosstalk between Notch, HIF-1alpha and GPER in Breast Cancer EMT. Int J Mol Sci. 2018;19:2011.

  57. Kazi AA, Gilani RA, Schech AJ, Chumsri S, Sabnis G, Shah P, et al. Nonhypoxic regulation and role of hypoxia-inducible factor 1 in aromatase inhibitor resistant breast cancer. Breast Cancer Res. 2014;16:R15.

    Article  Google Scholar 

  58. Wang KK, Yang Z, Sarkis G, Torres I, Raghavan V. Ubiquitin C-terminal hydrolase-L1 (UCH-L1) as a therapeutic and diagnostic target in neurodegeneration, neurotrauma and neuro-injuries. Expert Opin Ther Targets. 2017;21:627–38.

    Article  CAS  Google Scholar 

  59. Schroder C, Milde-Langosch K, Gebauer F, Schmid K, Mueller V, Wirtz RM, et al. Prognostic relevance of ubiquitin C-terminal hydrolase L1 (UCH-L1) mRNA and protein expression in breast cancer patients. J Cancer Res Clin Oncol. 2013;139:1745–55.

    Article  Google Scholar 

  60. Wang W, Zou L, Zhou D, Zhou Z, Tang F, Xu Z, et al. Overexpression of ubiquitin carboxyl terminal hydrolase-L1 enhances multidrug resistance and invasion/metastasis in breast cancer by activating the MAPK/Erk signaling pathway. Mol Carcinog. 2016;55:1329–42.

    Article  CAS  Google Scholar 

  61. Ning K, Wang T, Sun X, Zhang P, Chen Y, Jin J, et al. UCH-L1-containing exosomes mediate chemotherapeutic resistance transfer in breast cancer. J Surg Oncol. 2017;115:932–40.

    Article  CAS  Google Scholar 

  62. Chen XS, Wang KS, Guo W, Li LY, Yu P, Sun XY, et al. UCH-L1-mediated Down-regulation of Estrogen Receptor alpha Contributes to Insensitivity to Endocrine Therapy for Breast Cancer. Theranostics. 2020;10:1833–48.

    Article  CAS  Google Scholar 

  63. De Francesco EM, Sims AH, Maggiolini M, Sotgia F, Lisanti MP, Clarke RB. GPER mediates the angiocrine actions induced by IGF1 through the HIF-1alpha/VEGF pathway in the breast tumor microenvironment. Breast Cancer Res. 2017;19:129.

    Article  Google Scholar 

  64. De Francesco EM, Lappano R, Santolla MF, Marsico S, Caruso A, Maggiolini M. HIF-1alpha/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs). Breast Cancer Res. 2013;15:R64.

    Article  Google Scholar 

  65. Lappano R, Talia M, Cirillo F, Rigiracciolo DC, Scordamaglia D, Guzzi R, et al. The IL1beta-IL1R signaling is involved in the stimulatory effects triggered by hypoxia in breast cancer cells and cancer-associated fibroblasts (CAFs). J Exp Clin Cancer Res. 2020;39:153.

    Article  CAS  Google Scholar 

  66. Recchia AG, De Francesco EM, Vivacqua A, Sisci D, Panno ML, Ando S, et al. The G protein-coupled receptor 30 is up-regulated by hypoxia-inducible factor-1alpha (HIF-1alpha) in breast cancer cells and cardiomyocytes. J Biol Chem. 2011;286:10773–82.

    Article  CAS  Google Scholar 

  67. Győrffy B. Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer. Computational Struct Biotechnol J. 2021;19:4101–9.

    Article  Google Scholar 

Download references

Acknowledgements

This work was financially supported by the National Natural Science Foundation of China (81860648, 81973341), the Excellent Young Talents Plan of Guizhou Province (QKHPTRC(2021)-5632), the Guizhou Provincial Science and Technology Projects (QKHJC-ZK(2021)-YB527), the Excellent Young Talents Plan of Guizhou Medical University (2020-101), the Science and Technology Innovation Fund for Advanced Individuals of the Guizhou Department of Education (QJHKY[2018]048), and the Science Foundation of Administration of Traditional Chinese Medicine of Guizhou Province (QZYY-2022-025).

Author information

Authors and Affiliations

Authors

Contributions

YZ, YxS, SR and MqZ were responsible for designing the protocols, performing the experiments and writing the manuscript. ZZ was responsible for extracting and analysing data. SqF contributed to performing the experiments. XL contributed to interpreting results. XyP contributed to writing and revising the manuscript. QQ was responsible for drafting and revising the manuscript. XcS was responsible for approving the final version. YC was responsible for conceiving and designing the work that led to the submission, acquired data, played an important role in interpreting the results, and provided the funding resources.

Corresponding authors

Correspondence to Qi Qi, Xiangchun Shen or Yan Chen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Y., Song, Y., Ren, S. et al. GPER-mediated stabilization of HIF-1α contributes to upregulated aerobic glycolysis in tamoxifen-resistant cells. Oncogene 42, 184–197 (2023). https://doi.org/10.1038/s41388-022-02506-4

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02506-4

This article is cited by

Search

Quick links