Skip to content
2000
image of M1 Macrophage-derived TNF-α Promotes Pancreatic Cancer Ferroptosis Via p38 MAPK-ACSL4 Pathway

Abstract

Introduction

Pancreatic Ductal Adenocarcinoma (PDAC) is one of the most malignant gastrointestinal tumors. M1 macrophage, a subtype within the Tumor Microenvironment (TME), plays a vital role in the development of cancer. Despite its anti-tumoral functions, the specific mechanisms of its action remain incompletely understood.

Methods

The effect of M1 macrophages on the proliferation ability and cell viability of PDAC cells was evaluated by Cell Counting Kit-8 (CCK-8) cell proliferation assay, cell clone formation assay, and flow cytometry. Western blot, qRT-PCR, confocal microscope, RNA-sequencing, and transmission electron microscope were performed to assess lipid peroxidation and ferroptosis level of PDAC cells in the context of M1 macrophage or TNF-α.

Results

M1 macrophages inhibited cell proliferation and promoted cell death of PDAC cells, in which ferroptosis played a vital role. Mechanistically, Tumor Necrosis Factor-alpha (TNF-α) released by M1 macrophages binds to the TNFR1 receptor on pancreatic cancer cells, activating the p38 MAPK signaling, which upregulates Acyl-CoA Synthetase Long-chain family member 4 (ACSL4) expression, a critical lipid metabolism enzyme linked to ferroptosis, thereby promoting ferroptosis. Knockdown of ACSL4 or TNFR1 significantly reduced TNF-α-induced ferroptosis.

Discussion

TNF-α is a major inflammatory cytokine and is mainly generated by macrophages and T lymphocytes. It is involved in many pathological processes, such as inflammatory diseases, autoimmune diseases, and cancer. Studies have shown that the administration of recombinant TNF-α can induce tumor regression in mice with sarcomas. In our study, systemic injection of TNF-α slowed the tumor growth in nude mice, but with no significant difference compared with the control group, which may partially be attributed to its angiogenic activity. TNF-α signals via two distinct membrane-binding receptors, TNFR1 and TNFR2, which regulate various diseases. In pancreatic cancer, the role of TNF-α is complex and poorly understood. In a previous study, they found that exogenous systemic administration of human TNF-α, which interacted with murine TNFR1, significantly increased overall tumor growth in the Panc02-PDAC model. Intriguingly, the loss of TNFR1 led to an impediment of immune cell infiltration into the tumor and impaired immunosurveillance, which accelerated tumor growth. This suggests that TNFR1 exerts both pro-tumoral and anti-tumoral functions in the Panc02-PDAC model, but the overall outcome is likely dependent on the spatiotemporal availability of TNF-α. However, systemic TNF-α injection can lead to severe side effects in animals, limiting its further application. In a recent study, TNFR2 was found to promote tumorigenesis and progression in the KPC-PDAC model. Knockdown of TNFR2 or pretreatment with an anti-TNFR2 antibody could significantly slow the tumor progression and incidence. In our study, TNFR2 was found to have a low expression in pancreatic cancer cells and was barely detected with the failure of knockdown. However, the cell lines used in the former study were established from a KPC mouse model, while our experiments were conducted using human PDAC cell lines. Contrary findings are possible as cell lines originate from two different species. However, we will further investigate the mechanism of this difference.

Conclusion

In summary, this study revealed that M1 macrophages could induce ferroptosis in pancreatic cancer cells through secreting TNF-α, indicating a potential therapeutic option for PDAC.

Loading

Article metrics loading...

/content/journals/cmm/10.2174/0115665240374551250630075409
2025-07-10
2025-09-14
Loading full text...

Full text loading...

References

  1. Siegel R.L. Giaquinto A.N. Jemal A. Cancer statistics, 2024. CA Cancer J. Clin. 2024 74 1 12 49 10.3322/caac.21820 38230766
    [Google Scholar]
  2. Macchini M. Centonze F. Peretti U. Treatment opportunities and future perspectives for pancreatic cancer patients with germline BRCA1-2 pathogenic variants. Cancer Treat. Rev. 2021 100 102262 10.1016/j.ctrv.2021.102262 34418781
    [Google Scholar]
  3. Cao D. Song Q. Li J. Jiang Y. Wang Z. Lu S. Opportunities and challenges in targeted therapy and immunotherapy for pancreatic cancer. Expert Rev. Mol. Med. 2021 23 e21 10.1017/erm.2021.26 34906271
    [Google Scholar]
  4. Stockwell B.R. Friedmann Angeli J.P. Bayir H. Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017 171 2 273 285 10.1016/j.cell.2017.09.021 28985560
    [Google Scholar]
  5. Lei G. Zhang Y. Koppula P. The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 2020 30 2 146 162 10.1038/s41422‑019‑0263‑3 31949285
    [Google Scholar]
  6. Chen X. Kang R. Kroemer G. Tang D. Ferroptosis in infection, inflammation, and immunity. J. Exp. Med. 2021 218 6 e20210518 10.1084/jem.20210518 33978684
    [Google Scholar]
  7. Xu H. Ye D. Ren M. Zhang H. Bi F. Ferroptosis in the tumor microenvironment: Perspectives for immunotherapy. Trends Mol. Med. 2021 27 9 856 867 10.1016/j.molmed.2021.06.014 34312075
    [Google Scholar]
  8. Lei G. Mao C. Yan Y. Zhuang L. Gan B. Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell 2021 12 11 836 857 10.1007/s13238‑021‑00841‑y 33891303
    [Google Scholar]
  9. Chen X. Kang R. Kroemer G. Tang D. Broadening horizons: The role of ferroptosis in cancer. Nat. Rev. Clin. Oncol. 2021 18 5 280 296 10.1038/s41571‑020‑00462‑0 33514910
    [Google Scholar]
  10. Di Somma S. Napolitano F. Portella G. Malfitano A.M. Cross talk of macrophages with tumor microenvironment cells and modulation of macrophages in cancer by virotherapy. Biomedicines 2021 9 10 1309 10.3390/biomedicines9101309 34680425
    [Google Scholar]
  11. Zhang H. Deng T. Liu R. CAF secreted miR-522 suppresses ferroptosis and promotes acquired chemo-resistance in gastric cancer. Mol. Cancer 2020 19 1 43 10.1186/s12943‑020‑01168‑8 32106859
    [Google Scholar]
  12. Wang W. Green M. Choi J.E. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019 569 7755 270 274 10.1038/s41586‑019‑1170‑y 31043744
    [Google Scholar]
  13. Yee P.P. Wei Y. Kim S.Y. Neutrophil-induced ferroptosis promotes tumor necrosis in glioblastoma progression. Nat. Commun. 2020 11 1 5424 10.1038/s41467‑020‑19193‑y 33110073
    [Google Scholar]
  14. Xu T. Yu S. Zhang J. Wu S. Dysregulated tumor-associated macrophages in carcinogenesis, progression and targeted therapy of gynecological and breast cancers. J. Hematol. Oncol. 2021 14 1 181 10.1186/s13045‑021‑01198‑9 34717710
    [Google Scholar]
  15. Gordon S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003 3 1 23 35 10.1038/nri978 12511873
    [Google Scholar]
  16. Yang X. Liu H. Ye T. AhR activation attenuates calcium oxalate nephrocalcinosis by diminishing M1 macrophage polarization and promoting M2 macrophage polarization. Theranostics 2020 10 26 12011 12025 10.7150/thno.51144 33204326
    [Google Scholar]
  17. Okada A. Yasui T. Hamamoto S. A review of prognostic and predictive biomarkers in breast cancer. Clin. Exp. Med. 2023 23 1 1 16 10.1007/s10238‑021‑00781‑2
    [Google Scholar]
  18. Park M.W. Cha H.W. Kim J. NOX4 promotes ferroptosis of astrocytes by oxidative stress-induced lipid peroxidation via the impairment of mitochondrial metabolism in Alzheimer’s diseases. Redox Biol. 2021 41 101947 10.1016/j.redox.2021.101947 33774476
    [Google Scholar]
  19. Tabnak P. HajiEsmailPoor Z, Soraneh S. Ferroptosis in lung cancer: From molecular mechanisms to prognostic and therapeutic opportunities. Front. Oncol. 2021 11 792827 10.3389/fonc.2021.792827 34926310
    [Google Scholar]
  20. Chen X. Li J. Kang R. Klionsky D.J. Tang D. Ferroptosis: Machinery and regulation. Autophagy 2021 17 9 2054 2081 10.1080/15548627.2020.1810918 32804006
    [Google Scholar]
  21. Qin H. Holdbrooks A.T. Liu Y. Reynolds S.L. Yanagisawa L.L. Benveniste E.N. SOCS3 deficiency promotes M1 macrophage polarization and inflammation. J. Immunol. 2012 189 7 3439 3448 10.4049/jimmunol.1201168 22925925
    [Google Scholar]
  22. Amano M.T. Castoldi A. Andrade-Oliveira V. The lack of PI3Kγ favors M1 macrophage polarization and does not prevent kidney diseases progression. Int. Immunopharmacol. 2018 64 151 161 10.1016/j.intimp.2018.08.020 30176533
    [Google Scholar]
  23. Moradi-Chaleshtori M. Bandehpour M. Heidari N. Mohammadi-Yeganeh S. Mahmoud Hashemi S. Exosome-mediated miR-33 transfer induces M1 polarization in mouse macrophages and exerts antitumor effect in 4T1 breast cancer cell line. Int. Immunopharmacol. 2021 90 107198 10.1016/j.intimp.2020.107198 33249048
    [Google Scholar]
  24. Mohapatra S. Pioppini C. Ozpolat B. Calin G.A. Non-coding RNAs regulation of macrophage polarization in cancer. Mol. Cancer 2021 20 1 24 10.1186/s12943‑021‑01313‑x 33522932
    [Google Scholar]
  25. Kitaura H. Marahleh A. Ohori F. Role of the interaction of tumor necrosis Factor-α and tumor necrosis factor receptors 1 and 2 in bone-related cells. Int. J. Mol. Sci. 2022 23 3 1481 10.3390/ijms23031481 35163403
    [Google Scholar]
  26. Taniuchi K. Furihata M. Hanazaki K. Peroxiredoxin 1 promotes pancreatic cancer cell invasion by modulating p38 MAPK activity. Pancreas 2015 44 2 331 340 10.1097/MPA.0000000000000270 25426613
    [Google Scholar]
  27. Korc M. p38 MAPK in pancreatic cancer: Finding a protective needle in the haystack. Clin. Cancer Res. 2014 20 23 5866 5868 10.1158/1078‑0432.CCR‑14‑1543 25135483
    [Google Scholar]
  28. Zhang Z. Chen W. Zhang S. Isoliquiritigenin inhibits pancreatic cancer progression through blockade of p38 MAPK-regulated autophagy. Phytomedicine 2022 106 154406 10.1016/j.phymed.2022.154406 36029643
    [Google Scholar]
  29. Qiu X. Shi Q. Zhang X. Shi X. Jiang H. Qin S. LncRNA A2M-AS1 promotes ferroptosis in pancreatic cancer via interacting with PCBP3. Mol. Cancer Res. 2022 20 11 1636 1645 10.1158/1541‑7786.MCR‑22‑0024 35920801
    [Google Scholar]
  30. Feng H. Stockwell B.R. Unsolved mysteries: How does lipid peroxidation cause ferroptosis? PLoS Biol. 2018 16 5 e2006203 10.1371/journal.pbio.2006203 29795546
    [Google Scholar]
  31. Hadian K. Stockwell B.R. SnapShot. Ferroptosis. Cell 2020 181 5 1188 1188.e1 10.1016/j.cell.2020.04.039 32470402
    [Google Scholar]
  32. Domagała-Haduch M. Gorzelak-Magiera A. Michalecki Ł. Gisterek-Grocholska I. Radiochemotherapy in pancreatic cancer. Curr. Oncol. 2024 31 6 3291 3300 10.3390/curroncol31060250 38920733
    [Google Scholar]
  33. Farhangnia P. Khorramdelazad H. Nickho H. Delbandi A.A. Current and future immunotherapeutic approaches in pancreatic cancer treatment. J. Hematol. Oncol. 2024 17 1 40 10.1186/s13045‑024‑01561‑6 38835055
    [Google Scholar]
  34. Singh G. Kutcher D. Lally R. Rai V. Targeting neoantigens in pancreatic ductal adenocarcinoma. Cancers 2024 16 11 2101 10.3390/cancers16112101 38893220
    [Google Scholar]
  35. Mahadiuzzaman A.S.M. Dain Md Opo F.A. Alkarim S. Stem cell-based targeted therapy in pancreatic cancer: Current approaches and future prospects. Tissue Cell 2024 89 102449 10.1016/j.tice.2024.102449 38924893
    [Google Scholar]
  36. Lei G. Zhuang L. Gan B. Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer 2022 22 7 381 396 10.1038/s41568‑022‑00459‑0 35338310
    [Google Scholar]
  37. Zhao Y. Li M. Yao X. HCAR1/MCT1 regulates tumor ferroptosis through the lactate-mediated AMPK-SCD1 activity and its therapeutic implications. Cell Rep. 2020 33 10 108487 10.1016/j.celrep.2020.108487 33296645
    [Google Scholar]
  38. Liao P. Wang W. Wang W. CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell 2022 40 4 365 378.e6 10.1016/j.ccell.2022.02.003 35216678
    [Google Scholar]
  39. Liu L. Li Y. Cao D. SIRT3 inhibits gallbladder cancer by induction of AKT-dependent ferroptosis and blockade of epithelial-mesenchymal transition. Cancer Lett. 2021 510 93 104 10.1016/j.canlet.2021.04.007 33872694
    [Google Scholar]
  40. Zhang Y. Li S. Li F. Lv C. Yang Q. High-fat diet impairs ferroptosis and promotes cancer invasiveness via downregulating tumor suppressor ACSL4 in lung adenocarcinoma. Biol. Direct 2021 16 1 10 10.1186/s13062‑021‑00294‑7 34053456
    [Google Scholar]
  41. Chen J. Ding C. Chen Y. ACSL4 promotes hepatocellular carcinoma progression via c-Myc stability mediated by ERK/FBW7/c-Myc axis. Oncogenesis 2020 9 4 42 10.1038/s41389‑020‑0226‑z 32350243
    [Google Scholar]
  42. Balkwill F. Tumour necrosis factor and cancer. Nat. Rev. Cancer 2009 9 5 361 371 10.1038/nrc2628 19343034
    [Google Scholar]
  43. Jang D. Lee A.H. Shin H.Y. The role of Tumor Necrosis Factor Alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int. J. Mol. Sci. 2021 22 5 2719 10.3390/ijms22052719 33800290
    [Google Scholar]
  44. Horiuchi T. Mitoma H. Harashima S. Tsukamoto H. Shimoda T. Transmembrane TNF-: Structure, function and interaction with anti-TNF agents. Rheumatology (Oxford) 2010 49 7 1215 1228 10.1093/rheumatology/keq031 20194223
    [Google Scholar]
  45. Bradley J.R. TNF‐mediated inflammatory disease. J. Pathol. 2008 214 2 149 160 10.1002/path.2287 18161752
    [Google Scholar]
  46. Bianchi A. De Castro Silva I. Deshpande N.U. Cell-autonomous Cxcl1 sustains tolerogenic circuitries and stromal inflammation via neutrophil-derived tnf in pancreatic cancer. Cancer Discov. 2023 13 6 1428 1453 10.1158/2159‑8290.CD‑22‑1046 36946782
    [Google Scholar]
  47. Zidi I. Mestiri S. Bartegi A. Amor N.B. TNF-α and its inhibitors in cancer. Med. Oncol. 2010 27 2 185 198 10.1007/s12032‑009‑9190‑3 19277912
    [Google Scholar]
  48. North R.J. Havell E.A. The antitumor function of tumor necrosis factor (TNF) II. Analysis of the role of endogenous TNF in endotoxin-induced hemorrhagic necrosis and regression of an established sarcoma. J. Exp. Med. 1988 167 3 1086 1099 10.1084/jem.167.3.1086 3258350
    [Google Scholar]
  49. Havell E.A. Fiers W. North R.J. The antitumor function of tumor necrosis factor (TNF), I. Therapeutic action of TNF against an established murine sarcoma is indirect, immunologically dependent, and limited by severe toxicity. J. Exp. Med. 1988 167 3 1067 1085 10.1084/jem.167.3.1067 3351434
    [Google Scholar]
  50. Siegmund D. Wajant H. TNF and TNF receptors as therapeutic targets for rheumatic diseases and beyond. Nat. Rev. Rheumatol. 2023 19 9 576 591 10.1038/s41584‑023‑01002‑7 37542139
    [Google Scholar]
  51. Shi G. Hu Y. TNFR1 and TNFR2, which link NF-κB activation, drive lung cancer progression, cell dedifferen-tiation, and metastasis. Cancers 2023 15 17 4299 10.3390/cancers15174299
    [Google Scholar]
  52. Chopra M. Lang I. Salzmann S. Tumor necrosis factor induces tumor promoting and anti-tumoral effects on pancreatic cancer via TNFR1. PLoS One 2013 8 9 e75737 10.1371/journal.pone.0075737 24098720
    [Google Scholar]
  53. Zhang X. Lao M. Xu J. Combination cancer immunotherapy targeting TNFR2 and PD-1/PD-L1 signaling reduces immunosuppressive effects in the microenvironment of pancreatic tumors. J. Immunother. Cancer 2022 10 3 e003982 10.1136/jitc‑2021‑003982 35260434
    [Google Scholar]
/content/journals/cmm/10.2174/0115665240374551250630075409
Loading
/content/journals/cmm/10.2174/0115665240374551250630075409
Loading

Data & Media loading...

Supplements

Supplementary material is available on the publisher's website along with the published article.


  • Article Type:
    Research Article
Keywords: MAPK pathway ; M1 macrophage ; ACSL4 ; ferroptosis ; pancreatic cancer
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error
Please enter a valid_number test