Blockade of Arf1-mediated lipid metabolism in cancers promotes tumor infiltration of cytotoxic T cells via the LPE-PPARγ-NF-κB-CCL5 pathway

Na Wang, Tiange Yao, Chenfei Luo, Ling Sun, Yuetong Wang, Steven X. Hou

PDF(4930 KB)
PDF(4930 KB)
Life Metabolism ›› 2023, Vol. 2 ›› Issue (5) : 225-237. DOI: 10.1093/lifemeta/load036
Original Article

Blockade of Arf1-mediated lipid metabolism in cancers promotes tumor infiltration of cytotoxic T cells via the LPE-PPARγ-NF-κB-CCL5 pathway

Author information +
History +

Abstract

Tumor immunotherapy has achieved breakthroughs in a variety of tumors. However, the systemic absence of T cells in tumors and immunosuppressive tumor microenvironment so far limits the efficacy of immunotherapy to a small population of patients. Therefore, novel agents to increase T-cell tumor infiltration are urgently needed in the clinic. We recently found that inhibition of the ADP-ribosylation factor 1 (Arf1)-mediated lipid metabolism not only kills cancer stem cells (CSCs) but also elicits an anti-tumor immune response. In this study, we revealed a mechanism that targeting Arf1 promotes the infiltration of cytotoxic T lymphocytes (CTLs) into tumors through the C-C chemokine ligand 5 (CCL5)-C-C chemokine receptor type 5 (CCR5) pathway. We found that blockage of Arf1 induces the production of the unsaturated fatty acid (PE 18:1) that binds and sequestrates peroxisome proliferator-activated receptor-γ (PPARγ) from the PPARγ-nuclear factor-κB (NF-κB) cytoplasmic complex. The released NF-κB was then phosphorylated and translocated into the nucleus to regulate the transcription of chemokine CCL5. CCL5 promoted infiltration of CTLs for tumor regression. Furthermore, the combination of the Arf1 inhibitor and programmed cell death protein 1 (PD-1) blockade induced an even stronger anti-tumor immunity. Therefore, targeting Arf1 represents a novel anti-tumor immune approach by provoking T-cell tumor infiltration and may provide a new strategy for tumor immunotherapy.

Keywords

immune checkpoint blockade / cancer stem cells / cytotoxic T cells / PPARγ / CCL5-CCR5 pathway / Arf1

Cite this article

Download citation ▾
Na Wang, Tiange Yao, Chenfei Luo, Ling Sun, Yuetong Wang, Steven X. Hou. Blockade of Arf1-mediated lipid metabolism in cancers promotes tumor infiltration of cytotoxic T cells via the LPE-PPARγ-NF-κB-CCL5 pathway. Life Metabolism, 2023, 2(5): 225‒237 https://doi.org/10.1093/lifemeta/load036

References

[1]
Reck M, Rodríguez-Abreu D, Robinson AG et al. Pembrolizumab versus Chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med 2016;375:1823–33.
CrossRef Google scholar
[2]
Wolchok JD, Kluger H, Callahan MK et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 2013;369:122–33.
CrossRef Google scholar
[3]
Zaretsky JM, Garcia-Diaz A, Shin DS et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016;375:819–29.
CrossRef Google scholar
[4]
Bruni D, Angell HK, Galon J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat Rev Cancer 2020;20:662–80.
CrossRef Google scholar
[5]
Tumeh PC, Harview Christina L, Yearley JH et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71.
CrossRef Google scholar
[6]
Binnewies M, Roberts EW, Kersten K et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med 2018;24:541–50.
CrossRef Google scholar
[7]
Ganesh K, Massague J. TGF-β inhibition and immunotherapy: checkmate. Immunity 2018;48:626–8.
CrossRef Google scholar
[8]
Wellenstein MD, de Visser KE. Cancer-cell-intrinsic mechanisms shaping the tumor immune landscape. Immunity 2018;48:399–416.
CrossRef Google scholar
[9]
Nagarsheth N, Wicha MS, Zou W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol 2017;17:559–72.
CrossRef Google scholar
[10]
Jain S, Annett SL, Morgan MP et al. The cancer stem cell niche in ovarian cancer and its impact on immune surveillance. Int J Mol Sci 2021;22:4091.
CrossRef Google scholar
[11]
Baldominos P, Barbera-Mourelle A, Barreiro O et al. Quiescent cancer cells resist T cell attack by forming an immunosuppressive niche. Cell 2022;185:1694–708.e19.
CrossRef Google scholar
[12]
Kaczmarek B, Verbavatz JM, Jackson CL. GBF1 and Arf1 function in vesicular trafficking, lipid homeostasis and organelle dynamics. Biol Cell 2017;109:391–9.
CrossRef Google scholar
[13]
Donaldson JG, Jackson CL. ARF family G proteins and their regulators: roles in membrane transport, development and disease. Nat Rev Mol Cell Biol 2011;12:362–75.
CrossRef Google scholar
[14]
D’Souza-Schorey C, Chavrier P. ARF proteins: roles in membrane traffic and beyond. Nat Rev Mol Cell Biol 2006;7:347–58.
CrossRef Google scholar
[15]
Wang G, Xu J, Zhao J et al. Arf1-mediated lipid metabolism sustains cancer cells and its ablation induces anti-tumor immune responses in mice. Nat Commun 2020;11:220.
CrossRef Google scholar
[16]
Casalou C, Faustino A, Barral DC. Arf proteins in cancer cell migration. Small GTPases 2016;7:270–82.
CrossRef Google scholar
[17]
Lang L, Shay C, Zhao X et al. Combined targeting of Arf1 and Ras potentiates anticancer activity for prostate cancer therapeutics. J Exp Clin Cancer Res 2017;36:112.
CrossRef Google scholar
[18]
Lopez-Cotarelo P, Gomez-Moreira C, Criado-Garcia O et al. Beyond chemoattraction: multifunctionality of chemokine receptors in leukocytes. Trends Immunol 2017;38:927–41.
CrossRef Google scholar
[19]
Luther SA, Cyster JG. Chemokines as regulators of T cell differentiation. Nat Immunol 2001;2:102–7.
CrossRef Google scholar
[20]
Blanpain C, Buser R, Power CA et al. A chimeric MIP 1aRANTES protein demonstrates the use of different regions of the RANTES protein to bind and activate its receptors. J Leukoc Biol 2001;69:977–85.
CrossRef Google scholar
[21]
Aldinucci D, Borghese C, Casagrande N. The CCL5/CCR5 axis in cancer progression. Cancers (Basel) 2020;12:1765.
CrossRef Google scholar
[22]
Velasco-Velázquez M, Xolalpa W, Pestell RG et al. The potential to target CCL5/CCR5 in breast cancer. Expert Opin Ther Targets 2014;18:1265–75.
CrossRef Google scholar
[23]
Monsalve FA, Pyarasani RD, Delgado-Lopez F et al. Peroxisome proliferator-activated receptor targets for the treatment of metabolic diseases. Mediators Inflamm 2013;2013:549627.
CrossRef Google scholar
[24]
Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications—a review. Nutr J 2014;13:17.
CrossRef Google scholar
[25]
Christofides A, Konstantinidou E, Jani C et al. The role of peroxisome proliferator-activated receptors (PPAR) in immune responses. Metabolism 2021;114:154338.
CrossRef Google scholar
[26]
Marion-Letellier R, Savoye G, Ghosh S. Fatty acids, eicosanoids and PPAR gamma. Eur J Pharmacol 2016;785:44–9.
CrossRef Google scholar
[27]
Singh SR, Zeng X, Zhao J et al. The lipolysis pathway sustains normal and transformed stem cells in adult Drosophila. Nature 2016;538:109–13.
CrossRef Google scholar
[28]
Wang G, Yin W, Shin H et al. Neuronal accumulation of peroxidated lipids promotes demyelination and neurodegeneration through the activation of the microglial NLRP3 inflammasome. Nature Aging 2021;1:1024–37.
CrossRef Google scholar
[29]
Hammond VJ, Morgan AH, Lauder S et al. Novel keto-phospholipids are generated by monocytes and macrophages, detected in cystic fibrosis, and activate peroxisome proliferator-activated receptor-γ. J Biol Chem 2012;287:41651–66.
CrossRef Google scholar
[30]
Morgan AH, Dioszeghy V, Maskrey BH et al. Phosphatidylethanolamine-esterified eicosanoids in the mouse: tissue localization and inflammation-dependent formation in Th-2 disease. J Biol Chem 2009;284:21185–91.
CrossRef Google scholar
[31]
Werts C, le Bourhis L, Liu J et al. Nod1 and Nod2 induce CCL5/RANTES through the NF-κB pathway. Eur J Immunol 2007;37:2499–508.
CrossRef Google scholar
[32]
Li J, Eu JQ, Kong Li R et al. Targeting metabolism in cancer cells and the tumour microenvironment for cancer therapy. Molecules 2020;25:4831.
CrossRef Google scholar
[33]
Hirschhorn T, Stockwell BR. The development of the concept of ferroptosis. Free Radic Biol Med 2019;133:130–43.
CrossRef Google scholar
[34]
Stockwell BR, Jiang X. A physiological function for ferroptosis in tumor suppression by the immune system. Cell Metab 2019;30:14–5.
CrossRef Google scholar
[35]
Wang W, Green M, Choi JE et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature 2019;569:270–4.
CrossRef Google scholar
[36]
Liao P, Wang W, Wang W et al. CD8+ T cells and fatty acids orchestrate tumor ferroptosis and immunity via ACSL4. Cancer Cell 2022;40:365–78.e6.
CrossRef Google scholar
[37]
Garcia-Bates TM, Lehmann GM, Simpson-Haidaris PJ et al. Role of peroxisome proliferator-activated receptor gamma and its ligands in the treatment of hematological malignancies. PPAR Res 2008;2008:834612.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2023 The Author(s) 2023. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(4930 KB)

Accesses

Citations

Detail

Sections
Recommended

/