Glycerol-3-phosphate acyltransferase 3-mediated lipid droplets accumulation confers chemoresistance of colorectal cancer

Ying Wang, Caihua Xu, Xianfeng Yang, Xiaofei Liu, Zijian Guo, Xinyu Lin, Lihua Li, Zhaohui Huang

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (2) : e486. DOI: 10.1002/mco2.486
ORIGINAL ARTICLE

Glycerol-3-phosphate acyltransferase 3-mediated lipid droplets accumulation confers chemoresistance of colorectal cancer

Author information +
History +

Abstract

Colorectal cancer (CRC) is the third most common malignancy worldwide. It is well known that lipid metabolism reprogramming contributes to the tumor progression. However, the lipid metabolic alterations and potential remodeling mechanism underlying the chemoresistance of CRC remain largely unclear. In this study, we compared the gene expression profiles of chemoresistant versus control CRC cells from the GEO database and identified a key factor, Glycerol-3-phosphate acyltransferase 3 (GPAT3), that promotes lipid droplet (LD) production and confers chemoresistance of CRC. With applying of HPLC–MS and molecular dynamics simulation, we also demonstrated that the activity of lysophosphatidic acid synthesis by GPAT3 was dependent on its acetylation at K316 site. In particular, GPAT3-mediated LD accumulation inhibited immunogenic cell death of tumor, and thus facilitated CD8+ T-cell exhaustion and malignant progression in mouse xenografts and hepatic-metastasis tumors in CRC patients. High GPAT3 expression turned CRC cells into nonimmunogenic cells after (Oxaliplatin) Oxa treatment, which was supported by a decrease in cytotoxic IFN-γ release and CD8+ T-cell exhaustion. In conclusion, these findings revealed the role of GPAT3-associated LD accumulation, which conferred a malignant phenotype (chemoresistance) and regulated the tumor microenvironment of CRC. These results suggest that GPAT3 is a potential target to enhance CRC chemosensitivity and develop novel therapeutic interventions.

Cite this article

Download citation ▾
Ying Wang, Caihua Xu, Xianfeng Yang, Xiaofei Liu, Zijian Guo, Xinyu Lin, Lihua Li, Zhaohui Huang. Glycerol-3-phosphate acyltransferase 3-mediated lipid droplets accumulation confers chemoresistance of colorectal cancer. MedComm, 2024, 5(2): e486 https://doi.org/10.1002/mco2.486

References

[1]
Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209-249.
[2]
Bao MH, Hypoxia Wong CC. Metabolic reprogramming, and drug resistance in liver cancer. Cells. 2021;10(7):1715-1732.
[3]
Schiliro C, Firestein BL. Mechanisms of metabolic reprogramming in cancer cells supporting enhanced growth and proliferation. Cells. 2021;10(5):1056-1096.
[4]
Zhang Q, Zhou W, Yu S, et al. Metabolic reprogramming of ovarian cancer involves ACSL1-mediated metastasis stimulation through upregulated protein myristoylation. Oncogene. 2021;40(1):97-111.
[5]
Huang M, Koizumi A, Narita S, et al. Altering phosphoinositides in high-fat diet-associated prostate tumor xenograft growth. MedComm. 2021;2(4):756-764.
[6]
Wong A, Chen S, Yang LK, Kanagasundaram Y, Crasta K. Lipid accumulation facilitates mitotic slippage-induced adaptation to anti-mitotic drug treatment. Cell Death Discov. 2018;4:109-123.
[7]
Royo-Garcia A, Courtois S, Parejo-Alonso B, Espiau-Romera P, Sancho P. Lipid droplets as metabolic determinants for stemness and chemoresistance in cancer. World J Stem Cells. 2021;13(9):1307-1317.
[8]
Cadenas C, Vosbeck S, Edlund K, et al. LIPG-promoted lipid storage mediates adaptation to oxidative stress in breast cancer. Int J Cancer. 2019;145(4):901-915.
[9]
Curtarello M, Tognon M, Venturoli C, et al. Rewiring of lipid metabolism and storage in ovarian cancer cells after anti-VEGF therapy. Cells. 2019;8(12):1601-1619.
[10]
Tirinato L, Pagliari F, Di Franco S, et al. ROS and lipid droplet accumulation induced by high glucose exposure in healthy colon and colorectal cancer stem cells. Genes Dis. 2020;7(4):620-635.
[11]
Wu C, Dai C, Li X, et al. AKR1C3-dependent lipid droplet formation confers hepatocellular carcinoma cell adaptability to targeted therapy. Theranostics. 2022;12(18):7681-7698.
[12]
Tirinato L, Liberale C, Di Franco S, et al. Lipid droplets: a new player in colorectal cancer stem cells unveiled by spectroscopic imaging. Stem Cells. 2015;33(1):35-44.
[13]
Cotte AK, Aires V, Fredon M, et al. Lysophosphatidylcholine acyltransferase 2-mediated lipid droplet production supports colorectal cancer chemoresistance. Nat Commun. 2018;9(1):322-337.
[14]
Schlaepfer IR, Hitz CA, Gijon MA, Bergman BC, Eckel RH, Jacobsen BM. Progestin modulates the lipid profile and sensitivity of breast cancer cells to docetaxel. Mol Cell Endocrinol. 2012;363(1-2):111-121.
[15]
Montopoli M, Bellanda M, Lonardoni F, et al. Metabolic reprogramming” in ovarian cancer cells resistant to cisplatin. Curr Cancer Drug Targets. 2011;11(2):226-235.
[16]
Jin L, Zhu LY, Pan YL, Fu HQ, Zhang J. Prothymosin alpha promotes colorectal carcinoma chemoresistance through inducing lipid droplet accumulation. Mitochondrion. 2021;59:123-134.
[17]
Zhang X, Huang C, Yuan Y, et al. FOXM1-mediated activation of phospholipase D1 promotes lipid droplet accumulation and reduces ROS to support paclitaxel resistance in metastatic cancer cells. Free Radic Biol Med. 2022;179:213-228.
[18]
Birmpilis AI, Paschalis A, Mourkakis A, et al. Immunogenic cell death, DAMPs and prothymosin alpha as a putative anticancer immune response biomarker. Cells. 2022;11(9):1415-1435.
[19]
Tesniere A, Apetoh L, Ghiringhelli F, et al. Immunogenic cancer cell death: a key-lock paradigm. Curr Opin Immunol. 2008;20(5):504-511.
[20]
Sistigu A, Yamazaki T, Vacchelli E, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20(11):1301-1309.
[21]
Jensen NF, Stenvang J, Beck MK, et al. Establishment and characterization of models of chemotherapy resistance in colorectal cancer: towards a predictive signature of chemoresistance. Mol Oncol. 2015;9(6):1169-1185.
[22]
Wu Y, Chen K, Li L, et al. Plin2-mediated lipid droplet mobilization accelerates exit from pluripotency by lipidomic remodeling and histone acetylation. Cell Death Differ. 2022;29(11):2316-2331.
[23]
Alrob OA, Sankaralingam S, Ma C, et al. Obesity-induced lysine acetylation increases cardiac fatty acid oxidation and impairs insulin signalling. Cardiovasc Res. 2014;103(4):485-497.
[24]
Wang T, Chen K, Yao W, et al. Acetylation of lactate dehydrogenase B drives NAFLD progression by impairing lactate clearance. J Hepatol. 2021;74(5):1038-1052.
[25]
Zhao S, Xu W, Jiang W, et al. Regulation of cellular metabolism by protein lysine acetylation. Science. 2010;327(5968):1000-1004.
[26]
Jaime-Sanchez P, Uranga-Murillo I, Aguilo N, et al. Cell death induced by cytotoxic CD8(+) T cells is immunogenic and primes caspase-3-dependent spread immunity against endogenous tumor antigens. J Immunother Cancer. 2020;8(1):528-541.
[27]
Fuertes Marraco SA, Neubert NJ, Verdeil G, Speiser DE. Inhibitory receptors beyond T cell exhaustion. Front Immunol. 2015;6:310-323.
[28]
Asare-Werehene M, Communal L, Carmona E, et al. Plasma gelsolin inhibits CD8(+) T-cell function and regulates glutathione production to confer chemoresistance in ovarian cancer. Cancer Res. 2020;80(18):3959-3971.
[29]
Mehta A, Ratre YK, Soni VK, et al. Orchestral role of lipid metabolic reprogramming in T-cell malignancy. Front Oncol. 2023;13:1122789-1122807.
[30]
Han Y, Cai H, Ma L, et al. Nuclear orphan receptor NR4A2 confers chemoresistance and predicts unfavorable prognosis of colorectal carcinoma patients who received postoperative chemotherapy. Eur J Cancer. 2013;49(16):3420-3430.
[31]
Wang Y, Zhang H, Wang M, et al. CCNB2/SASP/Cathepsin B & PGE2 axis induce cell senescence mediated malignant transformation. Int J Biol Sci. 2021;17(13):3538-3553.
[32]
Liang J, Cao R, Wang X, et al. Mitochondrial PKM2 regulates oxidative stress-induced apoptosis by stabilizing Bcl2. Cell Res. 2017;27(3):329-351.
[33]
Zhang D, Shi R, Xiang W, et al. The Agpat4/LPA axis in colorectal cancer cells regulates antitumor responses via p38/p65 signaling in macrophages. Signal Transduct Target Ther. 2020;5(1):24-36.
[34]
Karlsson M, Zhang C, Mear L, et al. A single-cell type transcriptomics map of human tissues. Sci Adv. 2021;7(31):2169-2177.

RIGHTS & PERMISSIONS

2024 2024 The Authors. MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.
PDF

Accesses

Citations

Detail

Sections
Recommended

/