Abstract
Background
Fasting-mimicking diet (FMD) is a safe and effective strategy in clinical oncology via metabolically restricting tumour growth and remodelling the immunity. To date, few studies have investigated the impact of on tumour-associated macrophages (TAMs), which are a crucial component of immune cells in the tumour microenvironment. Fasting can induce the ubiquitin-proteasome system (UPS) to regulate intracellular protein turnover homoeostasis, while Nuclear Factor Erythroid 2-like 1 (NRF1; encoded by the gene Nfe2l1), which controls proteasome gene transcription, may potentially be induced by fasting. However, whether NRF1 is induced by FMD/fasting, and how NRF1-mediated protein turnover works on TAMs remain unknown. This study investigated the hypothesis that FMD activates the anti-tumour immunity of TAMs by ubiquitinated protein metabolism.
Methods
Subcutaneous MC38 tumour models were established in WT and myeloid-specific NRF1 knockout (Mye-NFE2L1-/-) C57BL/6 mice, treated with FMD alone or combined with intraperitoneal Trex1 inhibitor (Trex1-IN-1). TAMs were isolated from tumour tissues using CD11b+ magnetic bead sorting. In vitro, bone marrow-derived macrophages (BMDMs) were co-cultured with MC38 in fasting medium, with MC38 proliferation assessed by CCK8 assay. BMDM-derived TAMs (B-TAMs) were induced by MC38 supernatant under fasting conditions. IFNβ levels in serum and cell supernatant were measured by ELISA. RNA-seq was performed to compare WT and Mye-NFE2L1-/- BMDMs under fasting conditions. Protein levels of cGAS-Sting pathway components, ubiquitinated proteins, and nuclear NRF1 were analysed by Western blot, while Trex1 ubiquitination was assessed by Co-IP. qPCR quantified IFNβ-related gene expression and mitochondrial DNA (mtDNA) copy number. Trex1-mitochondria colocalization was examined by immunofluorescence, and Trex1-bound mtDNA levels were determined by ChIP-qPCR.
Results
FMD/fasting triggers interferon-β (IFNβ) secretion in TAMs, which is driven by protein metabolism. In TAMs with FMD, an initial accumulation of ubiquitinated proteins occurs concomitantly with the induction of NRF1 in response to fasting-induced energy stress, leading to the ubiquitin/proteasome-dependent proteolysis of the three prime repair exonuclease 1 (Trex1) through UPS. Such a process engages type I interferon responses, which derepress the cGAS-Sting-IFNβ axis to promote anti-tumour effects of TAMs. In the absence of NRF1, Trex1 accumulates due to impaired UPS and binds to mtDNA, disrupting cGAS sensing of mtDNA to inhibit IFNβ secretion in TAMs, which attenuates anti-tumour effects of FMD/fasting.
Conclusion
In this study, we revealed for the first time that FMD/fasting coordinates NRF1-UPS and Trex1/Sting-mediated type I interferon responses in TAMs that contribute to suppressing tumour growth.

Graphical abstract: FMD upregulates the entry of NRF1 into the TAM nucleus to promote gene expression of proteasome subunits, which induces the ubiquitin/proteasome-dependent proteolysis of Trex1, leading to derepression of the cGAS-Sting-IFNβ axis. On the other hand, FMD triggers increasing of mtDNA in TAMs, promoting the cGAS-Sting-IFNβ axis to release IFNβ. In myeloid NRF1 knockout TAMs, transcriptional levels of proteasome subunits are reduced, resulting in impaired proteolysis of Trex1 and its subsequent accumulation during fasting. Then, Trex1 binds to mtDNA, directing the inhibition of the cGAS-Sting-IFNβ axis and inhibiting IFNβ secretion of macrophages.
This is a preview of subscription content, access via your institution
Access options
Subscribe to this journal
Receive 24 print issues and online access
$259.00 per year
only $10.79 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout






Data availability
The RNA-seq generated from this study have been deposited in the National Genomics Data Center under the project accession PRJNA1234688. Any additional information required to reanalyse the data reported in this paper is available from the lead contact upon request.
References
Vernieri C, Fucà G, Ligorio F, Huber V, Vingiani A, Iannelli F, et al. Fasting-Mimicking Diet Is Safe and Reshapes Metabolism and Antitumor Immunity in Patients with Cancer. Cancer Discov. 2022;12:90–107.
Ligorio F, Vingiani A, Torelli T, Sposetti C, Drufuca L, Iannelli F, et al. Early downmodulation of tumor glycolysis predicts response to fasting-mimicking diet in triple-negative breast cancer patients. Cell Metab. 2025;37:330–344.e7.
de Groot S, Lugtenberg RT, Cohen D, Welters MJP, Ehsan I, Vreeswijk MPG, et al. Fasting mimicking diet as an adjunct to neoadjuvant chemotherapy for breast cancer in the multicentre randomized Phase 2 DIRECT trial. Nat Commun. 2020;11:3083.
Blaževitš O, Di Tano M, Longo VD. Fasting and fasting mimicking diets in cancer prevention and therapy. Trends Cancer. 2023;9:212–22.
Gonzalez CA, Riboli E. Diet and cancer prevention: where we are, where we are going. Nutr Cancer. 2006;56:225–31.
Kumar A, Chinnathambi S, Kumar M, Pandian GN. Food Intake and Colorectal Cancer. Nutr Cancer. 2023;75:1710–42.
Schmid A, Mathies V, Buentzel J, Keinki C, Huebner J. Diet Changes and Underlying Motives in Cancer Patients. Nutr Cancer. 2022;74:2017–28.
Brandhorst S, Levine ME, Wei M, Shelehchi M, Morgan TE, Nayak KS, et al. Fasting-mimicking diet causes hepatic and blood markers changes indicating reduced biological age and disease risk. Nat Commun. 2024;15:1309.
Zhong Z, Zhang H, Nan K, Zhong J, Wu Q, Lu L, et al. Fasting-Mimicking Diet Drives Antitumor Immunity against Colorectal Cancer by Reducing IgA-Producing Cells. Cancer Res. 2023;83:3529–43.
Nan K, Zhong Z, Yue Y, Shen Y, Zhang H, Wang Z, et al. Fasting-mimicking diet-enriched Bifidobacterium pseudolongum suppresses colorectal cancer by inducing memory CD8(+) T cells. Gut. 2025;74:775–86.
Di Biase S, Lee C, Brandhorst S, Manes B, Buono R, Cheng CW, et al. Fasting-Mimicking Diet Reduces HO-1 to Promote T Cell-Mediated Tumor Cytotoxicity. Cancer Cell. 2016;30:136–46.
Chen W, Zhou M, Guan B, Xie B, Liu Y, He J, et al. Tumour-associated macrophage-derived DOCK7-enriched extracellular vesicles drive tumour metastasis in colorectal cancer via the RAC1/ABCA1 axis. Clin Transl Med. 2024;14:e1591.
Gan S, Macalinao DG, Shahoei SH, Tian L, Jin X, Basnet H, et al. Distinct tumor architectures and microenvironments for the initiation of breast cancer metastasis in the brain. Cancer Cell. 2024;42:1693–1712.e24.
Sun P, Wang H, He Z, Chen X, Wu Q, Chen W, et al. Fasting inhibits colorectal cancer growth by reducing M2 polarization of tumor-associated macrophages. Oncotarget. 2017;8:74649–60.
Wang L, Wang YJ, Wang R, Gong FL, Shi YH, Li SN, et al. Fasting mimicking diet inhibits tumor-associated macrophage survival and pro-tumor function in hypoxia: implications for combination therapy with anti-angiogenic agent. J Transl Med. 2023;21:754.
Cassetta L, Pollard JW. A timeline of tumour-associated macrophage biology. Nat Rev Cancer. 2023;23:238–57.
Chen Y, Song Y, Du W, Gong L, Chang H, Zou Z. Tumor-associated macrophages: an accomplice in solid tumor progression. J Biomed Sci. 2019;26:78.
Lei A, Yu H, Lu S, Lu H, Ding X, Tan T, et al. A second-generation M1-polarized CAR macrophage with antitumor efficacy. Nat Immunol. 2024;25:102–16.
Xu M, Hu Y, Wu J, Liu J, Pu K. Sonodynamic Nano-LYTACs Reverse Tumor Immunosuppressive Microenvironment for Cancer Immunotherapy. J Am Chem Soc. 2024;146:34669–80.
Han S, Bao X, Zou Y, Wang L, Li Y, Yang L, et al. d-lactate modulates M2 tumor-associated macrophages and remodels immunosuppressive tumor microenvironment for hepatocellular carcinoma. Sci Adv. 2023;9:eadg2697.
Elewaut A, Estivill G, Bayerl F, Castillon L, Novatchkova M, Pottendorfer E, et al. Cancer cells impair monocyte-mediated T cell stimulation to evade immunity. Nature. 2025;637:716–25.
Sistigu A, Yamazaki T, Vacchelli E, Chaba K, Enot DP, Adam J, et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat Med. 2014;20:1301–9.
Borden EC. Interferons α and β in cancer: therapeutic opportunities from new insights. Nat Rev Drug Discov. 2019;18:219–34.
Dunn GP, Koebel CM, Schreiber RD. Interferons, immunity and cancer immunoediting. Nat Rev Immunol. 2006;6:836–48.
Pages F, Vives V, Sautès-Fridman C, Fossiez F, Berger A, Cugnenc PH, et al. Control of tumor development by intratumoral cytokines. Immunol Lett. 1999;68:135–9.
Qin A. An anti-cancer surveillance by the interplay between interferon-beta and retinoblastoma protein RB1. Front Oncol. 2023;13:1173467.
Triozzi P, Rinehart J. The role of IFN-beta in cancer therapy. Cancer Surv. 1989;8:799–808.
Maxwell MB, Hom-Tedla MS, Yi J, Li S, Rivera SA, Yu J, et al. ARID1A suppresses R-loop-mediated STING-type I interferon pathway activation of anti-tumor immunity. Cell. 2024;187:3390–3408.e19.
Yang CA, Huang HY, Chang YS, Lin CL, Lai IL, Chang JG. DNA-Sensing and Nuclease Gene Expressions as Markers for Colorectal Cancer Progression. Oncology. 2017;92:115–24.
An J, Woodward JJ, Lai W, Minie M, Sun X, Tanaka L, et al. Inhibition of Cyclic GMP-AMP Synthase Using a Novel Antimalarial Drug Derivative in Trex1-Deficient Mice. Arthritis Rheumatol. 2018;70:1807–19.
Coderch C, Arranz-Herrero J, Nistal-Villan E, de Pascual-Teresa B, Rius-Rocabert S. The Many Ways to Deal with STING. Int J Mol Sci. 2023;24.
Willemsen J, Neuhoff MT, Hoyler T, Noir E, Tessier C, Sarret S, et al. TNF leads to mtDNA release and cGAS/STING-dependent interferon responses that support inflammatory arthritis. Cell Rep. 2021;37:109977.
Ghosh M, Saha S, Li J, Montrose DC, Martinez LA. p53 engages the cGAS/STING cytosolic DNA sensing pathway for tumor suppression. Mol Cell. 2023;83:266–280.e6.
Mekers VE, Kho VM, Ansems M, Adema GJ. cGAS/cGAMP/STING signal propagation in the tumor microenvironment: Key role for myeloid cells in antitumor immunity. Radiother Oncol. 2022;174:158–67.
Oduro PK, Zheng X, Wei J, Yang Y, Wang Y, Zhang H, et al. The cGAS-STING signaling in cardiovascular and metabolic diseases: Future novel target option for pharmacotherapy. Acta Pharm Sin B. 2022;12:50–75.
Chen C, Xu P. Cellular functions of cGAS-STING signaling. Trends Cell Biol. 2023;33:630–48.
Tani T, Mathsyaraja H, Campisi M, Li ZH, Haratani K, Fahey CG, et al. TREX1 Inactivation Unleashes Cancer Cell STING-Interferon Signaling and Promotes Antitumor Immunity. Cancer Discov. 2024;14:752–65.
Zhou W, Richmond-Buccola D, Wang Q, Kranzusch PJ. Structural basis of human TREX1 DNA degradation and autoimmune disease. Nat Commun. 2022;13:4277.
Mohr L, Toufektchan E, von Morgen P, Chu K, Kapoor A, Maciejowski J. ER-directed TREX1 limits cGAS activation at micronuclei. Mol Cell. 2021;81:724–738.e9.
Xue Y, Wang Y, Ren Z, Yu K. Tissue factor promotes TREX1 protein stability to evade cGAS-STING innate immune response in pancreatic ductal adenocarcinoma. Oncogene. 2025;44:739–52.
Wang C, Zhang R, He J, Yu L, Li X, Zhang J, et al. Ultrasound-responsive low-dose doxorubicin liposomes trigger mitochondrial DNA release and activate cGAS-STING-mediated antitumour immunity. Nat Commun. 2023;14:3877.
Falahat R, Berglund A, Perez-Villarroel P, Putney RM, Hamaidi I, Kim S, et al. Epigenetic state determines the in vivo efficacy of STING agonist therapy. Nat Commun. 2023;14:1573.
Chakraborty S, Sen U, Ventura K, Jethalia V, Coleman C, Sridhar S, et al. Lurbinectedin sensitizes PD-L1 blockade therapy by activating STING-IFN signaling in small-cell lung cancer. Cell Rep Med. 2025;6:101944.
Nader GPF, Agüera-Gonzalez S, Routet F, Gratia M, Maurin M, Cancila V, et al. Compromised nuclear envelope integrity drives TREX1-dependent DNA damage and tumor cell invasion. Cell. 2021;184:5230–5246.e22.
Zhang W, Wang G, Xu ZG, Tu H, Hu F, Dai J, et al. Lactate Is a Natural Suppressor of RLR Signaling by Targeting MAVS. Cell. 2019;178:176–189.e15.
Tang J, Wang X, Chen S, Chang T, Gu Y, Zhang F, et al. Disruption of glucose homeostasis by bacterial infection orchestrates host innate immunity through NAD(+)/NADH balance. Cell Rep. 2024;43:114648.
Bi T, Liang P, Zhou Y, Wang H, Huang R, Sun Q, et al. Rational Design of Bioorthogonally Activatable PROTAC for Tumor-Targeted Protein Degradation. J Med Chem. 2023;66:14843–52.
Sun-Wang JL, Yarritu-Gallego A, Ivanova S, Zorzano A. The ubiquitin-proteasome system and autophagy: self-digestion for metabolic health. Trends Endocrinol Metab. 2021;32:594–608.
Wang T, Zhang Y, Liu Y, Huang Y, Wang W. Amino Acid-Starved Cancer Cells Utilize Macropinocytosis and Ubiquitin-Proteasome System for Nutrient Acquisition. Adv Sci. 2024;11:e2304791.
Yan J, Zhang X, Wang H, Jia X, Wang R, Wu S, et al. Macrophage NRF1 promotes mitochondrial protein turnover via the ubiquitin proteasome system to limit mitochondrial stress and inflammation. Cell Rep. 2024;43:114780.
Argüello RJ, Rodriguez Rodrigues C, Gatti E, Pierre P. Protein synthesis regulation, a pillar of strength for innate immunity?. Curr Opin Immunol. 2015;32:28–35.
Finn PF, Dice JF. Proteolytic and lipolytic responses to starvation. Nutrition. 2006;22:830–44.
Zhang Y, Xiang Y. Molecular and cellular basis for the unique functioning of Nrf1, an indispensable transcription factor for maintaining cell homoeostasis and organ integrity. Biochem J. 2016;473:961–1000.
Bartelt A, Widenmaier SB, Schlein C, Johann K, Goncalves RLS, Eguchi K, et al. Brown adipose tissue thermogenic adaptation requires Nrf1-mediated proteasomal activity. Nat Med. 2018;24:292–303.
Zhang Y, Manning BD. mTORC1 signaling activates NRF1 to increase cellular proteasome levels. Cell Cycle. 2015;14:2011–7.
Fu J, Zheng H, Cui Q, Chen C, Bao S, Sun J, et al. Nfe2l1-silenced insulinoma cells acquire aggressiveness and chemoresistance. Endocr Relat Cancer. 2018;25:185–200.
De I, Dogra N, Singh S. The mitochondrial unfolded protein response: role in cellular homeostasis and disease. Curr Mol Med. 2017;17:587–97.
Wang Q, Bergholz JS, Ding L, Lin Z, Kabraji SK, Hughes ME, et al. STING agonism reprograms tumor-associated macrophages and overcomes resistance to PARP inhibition in BRCA1-deficient models of breast cancer. Nat Commun. 2022;13:3022.
Byles V, Covarrubias AJ, Ben-Sahra I, Lamming DW, Sabatini DM, Manning BD, et al. The TSC-mTOR pathway regulates macrophage polarization. Nat Commun. 2013;4:2834.
Zhong W, Lu Y, Han X, Yang J, Qin Z, Zhang W, et al. Upregulation of exosome secretion from tumor-associated macrophages plays a key role in the suppression of anti-tumor immunity. Cell Rep. 2023;42:113224.
Salvadori G, Zanardi F, Iannelli F, Lobefaro R, Vernieri C, Longo VD. Fasting-mimicking diet blocks triple-negative breast cancer and cancer stem cell escape. Cell Metab. 2021;33:2247–2259.e6.
Liu P, Kerins MJ, Tian W, Neupane D, Zhang DD, Ooi A. Differential and overlapping targets of the transcriptional regulators NRF1, NRF2, and NRF3 in human cells. J Biol Chem. 2019;294:18131–49.
Blackwell TK, Steinbaugh MJ, Hourihan JM, Ewald CY, Isik M. SKN-1/Nrf, stress responses, and aging in Caenorhabditis elegans. Free Radic Biol Med. 2015;88:290–301.
Zhong S, Chen W, Wang B, Gao C, Liu X, Song Y, et al. Energy stress modulation of AMPK/FoxO3 signaling inhibits mitochondria-associated ferroptosis. Redox Biol. 2023;63:102760.
Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat Rev Mol Cell Biol. 2018;19:121–35.
Ronnebaum SM, Patterson C, Schisler JC. Minireview: hey U(PS): metabolic and proteolytic homeostasis linked via AMPK and the ubiquitin proteasome system. Mol Endocrinol. 2014;28:1602–15.
Cheng AN, Cheng LC, Kuo CL, Lo YK, Chou HY, Chen CH, et al. Mitochondrial Lon-induced mtDNA leakage contributes to PD-L1-mediated immunoescape via STING-IFN signaling and extracellular vesicles. J Immunother Cancer. 2020;8.
Sterman DH, Recio A, Haas AR, Vachani A, Katz SI, Gillespie CT, et al. A Phase I trial of repeated intrapleural adenoviral-mediated interferon-beta gene transfer for mesothelioma and metastatic pleural effusions. Mol Ther. 2010;18:852–60.
Hu X, Shang S, Nestorov I, Hasan J, Seddighzadeh A, Dawson K, et al. COMPARE: Pharmacokinetic profiles of subcutaneous peginterferon beta-1a and subcutaneous interferon beta-1a over 2 weeks in healthy subjects. Br J Clin Pharm. 2016;82:380–8.
King CR, Liu Y, Amato KA, Schaack GA, Mickelson C, Sanders AE, et al. Pathogen-driven CRISPR screens identify TREX1 as a regulator of DNA self-sensing during influenza virus infection. Cell Host Microbe. 2023;31:1552–1567.e8.
Anindya R. Cytoplasmic DNA in cancer cells: Several pathways that potentially limit DNase2 and TREX1 activities. Biochim Biophys Acta Mol Cell Res. 2022;1869:119278.
Caielli S, Cardenas J, de Jesus AA, Baisch J, Walters L, Blanck JP, et al. Erythroid mitochondrial retention triggers myeloid-dependent type I interferon in human SLE. Cell. 2021;184:4464–4479.e19.
Hu MM, Shu HB. Mitochondrial DNA-triggered innate immune response: mechanisms and diseases. Cell Mol Immunol. 2023;20:1403–12.
Zheng S, Song J, Linghu D, Yang R, Liu B, Xue Z, et al. Galectin-9 blockade synergizes with ATM inhibition to induce potent anti-tumor immunity. Int J Biol Sci. 2023;19:981–93.
Benci JL, Xu B, Qiu Y, Wu TJ, Dada H, Twyman-Saint Victor C, et al. Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade. Cell. 2016;167:1540–1554.e12.
López-Soto A, Gonzalez S, Folgueras AR. IFN Signaling and ICB Resistance: Time is on Tumor’s Side. Trends Cancer. 2017;3:161–3.
Acknowledgements
C.M. was supported by the Shanghai Clinical Research Center for Anaesthesiology (2023ZZ02001), the National Science and Technology Major Project of China (No2023YFA1800204), the Shanghai Municipal 2021 Science and Technology Innovation Action Plan (No.21JC1401400), the Shanghai Leading Talent (No. 2020-112), and the National Natural Science Foundation of China (NSFC, 82430068 and 82072213). T.H. was supported by funding from MOST (2021YFA0804700); NSFC (32070895, 92057105, 32225026); Foreign Experts Programs issued by Science & Technological Commission of Shanghai Municipality (STCSM, 22WZ2504000); and Shanghai Frontiers Science Center for Biomacromolecules and Precision Medicine at ShanghaiTech University and Shanghai Clinical Research and Trial Center, 201210, Shanghai, People’s Republic of China. Z.L. was supported by NSFC (82472212). W.J. was supported by funding from NSFC (32200715). Z.Z. was supported by funding from NSFC (82403441). We cordially thank Professor Tiffany Horng and Jiawei Yan from ShanghaiTech University for generating and providing mice with myeloid-specific knockdown of NRF1(NFE2L1).
Author information
Authors and Affiliations
Contributions
J.L. designed the project, performed the experiments, analysed the data and wrote the manuscript. C.M. supervised the project, including the experimental design and funding support. T.H. provided mice with a myeloid-specific knock of NRF1(NFE2L1) and experimental equipment support. Z.L. provided suggestions for the revised manuscript and funding support. W.J. provided suggestions for manuscript revision and contributed important insights about proteolysis and related technical expertise, including CoIP and ChIP. G.T. and Z.Z. engaged the animal experiments and provided the FMD protocol.
Corresponding authors
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.
About this article
Cite this article
Li, J., Jiang, W., Tu, G. et al. Fasting-mimicking diet induces IFNβ secretion in tumor-associated macrophages via NRF1-mediated ubiquitin-dependent proteolysis of Trex1. Br J Cancer (2026). https://doi.org/10.1038/s41416-025-03319-4
Received:
Revised:
Accepted:
Published:
Version of record:
DOI: https://doi.org/10.1038/s41416-025-03319-4