Skip to main content

Advertisement

Log in

G6PD activation in TNBC cells induces macrophage recruitment and M2 polarization to promote tumor progression

  • Original Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Glucose-6-phosphate dehydrogenase (G6PD) is involved in triple-negative breast cancer (TNBC) progression. Metabolic crosstalk between cancer cells and tumor-associated macrophages mediates tumor progression in TNBC. Molecular biological methods were applied to clarify the mechanism of the crosstalk between TNBC cells and M2 macrophages. In the present study, we verified that G6PD overexpression drives M2 macrophage polarization by directly combining with phospho-STAT1 and upregulating CCL2 and TGF-β1 secretion in TNBC cells. In turn, M2-like TAMs activated TNBC cells through IL-10 secretion, providing feedback to upregulate G6PD and promote TNBC cell migration and proliferation in vitro. Furthermore, we found that 6-AN (a specific inhibitor of G6PD) not only suppressed the cancer-driven polarization of macrophages toward the M2 phenotype but also inhibited the inherent M2 polarization of macrophages. Targeting the G6PD-regulated pentose phosphate pathway restrained TNBC progression and M2-type polarization of macrophages in vitro and in vivo.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data availability

The authors confirm that the data supporting the findings of this study are available within the article. The raw data are available from the corresponding author upon reasonable request. Figure 1D data analyzed in this study were obtained from the Gene Expression Omnibus (GEO) at GSE81032.

Abbreviations

TNBC:

Triple-negative breast cancer.

PPP:

Pentose phosphate pathway.

G6PD:

Glucose-6-phosphate dehydrogenase.

6-AN:

6-Aminonicotinamide.

TAMs:

Tumor-associated macrophages.

Mφ:

Macrophage.

ELISA:

Enzyme-linked Immunosorbent Assay.

CM:

Conditioned medium.

q-PCR:

Quantitative polymerase chain reaction.

MTT:

Thiazolyl blue.

IHC:

Immunohistochemistry.

H&E:

Hematoxylin and eosin.

DAPI:

4′,6-Diamidino-2-phenylindole.

CCL2:

Chemokine (C–C motif) ligand 2

TGF-β1:

Transforming growth factor beta 1

IL-10:

Interleukin-10

References

  1. Siegel RL et al (2021) Cancer Statistics, 2021. CA Cancer J Clin 71(1):7–33

    PubMed  Google Scholar 

  2. Yin L et al (2020) Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res 22(1):61

    PubMed  PubMed Central  Google Scholar 

  3. Robey IF et al (2005) Hypoxia-inducible factor-1alpha and the glycolytic phenotype in tumors. Neoplasia 7(4):324–330

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Pu H et al (2015) Overexpression of G6PD is associated with high risks of recurrent metastasis and poor progression-free survival in primary breast carcinoma. World J Surg Oncol 13:323

    PubMed  PubMed Central  Google Scholar 

  5. Dong T et al (2016) Altered glycometabolism affects both clinical features and prognosis of triple-negative and neoadjuvant chemotherapy-treated breast cancer. Tumour Biol 37(6):8159–8168

    CAS  PubMed  Google Scholar 

  6. Tu D et al (2021) M2 macrophages contribute to cell proliferation and migration of breast cancer. Cell Biol Int 45(4):831–838

    CAS  PubMed  Google Scholar 

  7. Jayasingam SD et al (2019) Evaluating the polarization of tumor-associated macrophages into m1 and m2 phenotypes in human cancer tissue: technicalities and challenges in routine clinical practice. Front Oncol 9:1512

    PubMed  Google Scholar 

  8. Chen P et al (2017) Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proc Natl Acad Sci USA 114(3):580–585

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Chen F et al (2019) Extracellular vesicle-packaged HIF-1alpha-stabilizing lncRNA from tumour-associated macrophages regulates aerobic glycolysis of breast cancer cells. Nat Cell Biol 21(4):498–510

    CAS  PubMed  Google Scholar 

  10. Li Y et al. (2022) Targeting glucose-6-phosphate dehydrogenase by 6-AN induces ROS-mediated autophagic cell death in breast cancer. FEBS J.

  11. Shi T et al. Conservation of protein abundance patterns reveals the regulatory architecture of the EGFR-MAPK pathway. Sci Signal, 2016. 9(436): p. rs6.

  12. Dias AS et al (2019) Metabolic crosstalk in the breast cancer microenvironment. Eur J Cancer 121:154–171

    CAS  PubMed  Google Scholar 

  13. Zheng H et al. (2021) Tumor Microenvironment: Key Players in Triple Negative Breast Cancer Immunomodulation. Cancers (Basel) 13(13).

  14. Nishida-Aoki N, Gujral TS (2019) Emerging approaches to study cell-cell interactions in tumor microenvironment. Oncotarget 10(7):785–797

    PubMed  PubMed Central  Google Scholar 

  15. Tymoszuk P et al (2014) High STAT1 mRNA levels but not its tyrosine phosphorylation are associated with macrophage infiltration and bad prognosis in breast cancer. BMC Cancer 14:257

    PubMed  PubMed Central  Google Scholar 

  16. Tilborghs S et al (2017) The role of Nuclear Factor-kappa B signaling in human cervical cancer. Crit Rev Oncol Hematol 120:141–150

    PubMed  Google Scholar 

  17. Ma X et al (2017) Polo-like kinase 1 coordinates biosynthesis during cell cycle progression by directly activating pentose phosphate pathway. Nat Commun 8(1):1506

    PubMed  PubMed Central  Google Scholar 

  18. Ni Y et al (2021) Silent information regulator 2 promotes clear cell renal cell carcinoma progression through deacetylation and small ubiquitin-related modifier 1 modification of glucose 6-phosphate dehydrogenase. Cancer Sci 112(10):4075–4086

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Butturini E, Carcereri de Prati A, Mariotto S (2020) Redox Regulation of STAT1 and STAT3 Signaling. Int J Mol Sci. 21(19).

  20. Verhoeven Y et al (2020) The potential and controversy of targeting STAT family members in cancer. Semin Cancer Biol 60:41–56

    CAS  PubMed  Google Scholar 

  21. Ch'ng ES, Jaafar H (2011) Tuan Sharif SE Breast Tumor Angiogenesis and Tumor-Associated Macrophages: Histopathologist's Perspective. Patholog Res Int. 2011: 572706.

  22. Wang H, et al. (2021) The impact of the tumor microenvironment on macrophage polarization in cancer metastatic progression. Int J Mol Sci. 22(12).

  23. Owen KL, Brockwell NK, Parker BS (2019) JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers (Basel). 11(12).

  24. Sun SC (2017) The non-canonical NF-kappaB pathway in immunity and inflammation. Nat Rev Immunol 17(9):545–558

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Sun L et al (2018) Metabolic reprogramming for cancer cells and their microenvironment: beyond the Warburg Effect. Biochim Biophys Acta Rev Cancer 1870(1):51–66

    CAS  PubMed  Google Scholar 

  26. Jiang P, Du W, Wu M (2014) Regulation of the pentose phosphate pathway in cancer. Protein Cell 5(8):592–602

    CAS  PubMed  PubMed Central  Google Scholar 

  27. More TH et al (2018) Metabolomic alterations in invasive ductal carcinoma of breast: a comprehensive metabolomic study using tissue and serum samples. Oncotarget 9(2):2678–2696

    PubMed  Google Scholar 

  28. Tayyari F et al (2018) Metabolic profiles of triple-negative and luminal A breast cancer subtypes in African-American identify key metabolic differences. Oncotarget 9(14):11677–11690

    PubMed  PubMed Central  Google Scholar 

  29. Budczies J et al (2012) Remodeling of central metabolism in invasive breast cancer compared to normal breast tissue—a GC-TOFMS based metabolomics study. BMC Genomics 13:334

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Willmann L et al (2015) Metabolic profiling of breast cancer: differences in central metabolism between subtypes of breast cancer cell lines. J Chromatogr B Analyt Technol Biomed Life Sci 1000:95–104

    CAS  PubMed  Google Scholar 

  31. Tang X et al (2014) A joint analysis of metabolomics and genetics of breast cancer. Breast Cancer Res 16(4):415

    PubMed  PubMed Central  Google Scholar 

  32. Willmann L et al (2016) Alterations of the exo- and endometabolite profiles in breast cancer cell lines: a mass spectrometry-based metabolomics approach. Anal Chim Acta 925:34–42

    CAS  PubMed  Google Scholar 

  33. Patra KC, Hay N (2014) The pentose phosphate pathway and cancer. Trends Biochem Sci 39(8):347–354

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Yin X et al (2017) ID1 promotes hepatocellular carcinoma proliferation and confers chemoresistance to oxaliplatin by activating pentose phosphate pathway. J Exp Clin Cancer Res 36(1):166

    PubMed  PubMed Central  Google Scholar 

  35. Barajas JM et al (2018) The role of miR-122 in the dysregulation of glucose-6-phosphate dehydrogenase (G6PD) expression in hepatocellular cancer. Sci Rep 8(1):9105

    PubMed  PubMed Central  Google Scholar 

  36. Wu S et al (2018) Transcription factor YY1 promotes cell proliferation by directly activating the pentose phosphate pathway. Cancer Res 78(16):4549–4562

    CAS  PubMed  Google Scholar 

  37. Massari F et al (2016) Metabolic phenotype of bladder cancer. Cancer Treat Rev 45:46–57

    CAS  PubMed  Google Scholar 

  38. Pathria P, Louis TL, Varner JA (2019) Targeting tumor-associated macrophages in cancer. Trends Immunol 40(4):310–327

    CAS  PubMed  Google Scholar 

  39. Mantovani A et al (2017) Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 14(7):399–416

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Cook J, Hagemann T (2013) Tumour-associated macrophages and cancer. Curr Opin Pharmacol 13(4):595–601

    CAS  PubMed  Google Scholar 

  41. Pitt JM et al (2016) Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol 27(8):1482–1492

    CAS  PubMed  Google Scholar 

  42. Castellaro AM et al. (2019) Tumor-associated macrophages induce endocrine therapy resistance in ER+ breast cancer cells. Cancers (Basel). 11(2).

  43. Munoz-Garcia J et al (2021) The twin cytokines interleukin-34 and CSF-1: masterful conductors of macrophage homeostasis. Theranostics 11(4):1568–1593

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Sica A, Mantovani A (2012) Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 122(3):787–795

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Shapouri-Moghaddam A et al (2018) Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol 233(9):6425–6440

    CAS  PubMed  Google Scholar 

  46. Hu T et al (2013) Variant G6PD levels promote tumor cell proliferation or apoptosis via the STAT3/5 pathway in the human melanoma xenograft mouse model. BMC Cancer 13:251

    PubMed  PubMed Central  Google Scholar 

  47. Yang HC et al (2015) Glucose 6-phosphate dehydrogenase knockdown enhances IL-8 expression in HepG2 cells via oxidative stress and NF-kappaB signaling pathway. J Inflamm (Lond) 12:34

    PubMed  Google Scholar 

  48. Dai L et al (2019) SARI attenuates colon inflammation by promoting STAT1 degradation in intestinal epithelial cells. Mucosal Immunol 12(5):1130–1140

    CAS  PubMed  Google Scholar 

  49. Jia P et al. (2022) Chemokine CCL2 from proximal tubular epithelial cells contributes to sepsis-induced acute kidney injury. Am J Physiol Renal Physiol.

  50. Cao J et al (2021) NR4A1 knockdown confers hepatoprotection against ischaemia-reperfusion injury by suppressing TGFbeta1 via inhibition of CYR61/NF-kappaB in mouse hepatocytes. J Cell Mol Med 25(11):5099–5112

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Duke KS et al (2017) STAT1-dependent and -independent pulmonary allergic and fibrogenic responses in mice after exposure to tangled versus rod-like multi-walled carbon nanotubes. Part Fibre Toxicol 14(1):26

    PubMed  PubMed Central  Google Scholar 

  52. Zhang F et al (2016) TGF-beta induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget 7(32):52294–52306

    PubMed  PubMed Central  Google Scholar 

  53. Van den Bossche J, O’Neill LA, Menon D (2017) Macrophage Immunometabolism: Where Are We (Going)? Trends Immunol 38(6):395–406

    PubMed  Google Scholar 

  54. Noy R, Pollard JW (2014) Tumor-associated macrophages: from mechanisms to therapy. Immunity 41(1):49–61

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Ungefroren H et al. (2021) Autocrine TGFbeta1 opposes exogenous tgfbeta1-induced cell migration and growth arrest through sustainment of a feed-forward loop involving MEK-ERK signaling. Cancers (Basel), 13(6).

  56. David CJ et al (2016) TGF-beta tumor suppression through a Lethal EMT. Cell 164(5):1015–1030

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Chen RH et al (2020) Tumor cell-secreted ISG15 promotes tumor cell migration and immune suppression by inducing the macrophage M2-like phenotype. Front Immunol 11:594775

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Su B et al (2021) Let-7d inhibits intratumoral macrophage M2 polarization and subsequent tumor angiogenesis by targeting IL-13 and IL-10. Cancer Immunol Immunother 70(6):1619–1634

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank Gang Zhao (Department of Pathology, Tianjin Cancer Hospital) and the breast surgery department of Peking Union Medical College Hospital for supplying breast cancer tissue sections, Miaomiao Sheng (Kunming University of Science and Technology) for providing breast cancer cell lines, and Xinying Wu (Animal Center of Nankai University) for technical support of in vivo imaging.

Funding

This study was partially funded by “State Key Laboratory of Medicinal Chemical Biology (NanKai University, No. 2019014)” to C W and “The Fundamental Research Funds for the Central Universities (Nankai University, #ZB19100128)” to C L.

Author information

Authors and Affiliations

Authors

Contributions

Y L, X H and Z F designed, performed and analyzed the in vivo experiments. Y L, X H and Z F performed and analyzed G6PD knockdown experiments in vitro and in vivo. Y L, X H and Z L provided guidance on data processing and writing. C L, Y L, C W, Q S and Z F contributed to the study design, implementation and supervision of the study. Z L, Z F, C W, Q S and C L contributed to the study design, supervised the study, and reviewed the manuscript. Y L and C L wrote the manuscript. All authors had full access to the data and approved the final version of the manuscript.

Corresponding authors

Correspondence to Changjun Wang, Zhenkun Fu, Qiang Sun or Chenggang Li.

Ethics declarations

Conflict of interests

The authors declare that they have no competing interests.

Ethics approval and consent to participate

Animal experiments were performed according to the Guidelines on Laboratory Animals of Nankai University and were approved by the Institute Research Ethics Committee at Nankai University (No: 2021-SYDWLL-000464). Human experiments were approved by the human experimentation committee, and informed consent was obtained from all subjects (No: NKUIRB2021105).

Consent for publication

Not applicable.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 2164 KB)

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

Li, Y., Han, X., Lin, Z. et al. G6PD activation in TNBC cells induces macrophage recruitment and M2 polarization to promote tumor progression. Cell. Mol. Life Sci. 80, 165 (2023). https://doi.org/10.1007/s00018-023-04810-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00018-023-04810-y

Keywords

Navigation