AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (28.9 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Open Access

Intermittent fasting boosts antitumor immunity by restricting CD11b+Ly6Clow Ly6Glow cell viability through glucose metabolism in murine breast tumor model

Chenghao Fua,#Zhehao Lianga,#,Zemiao NiuaNing ChenaYuemin LiaZhenhua LiangaYanwei HuoaHao XiaRong WangaYonghuan Yanb,cXiaoru GanoaMengtian Wangb,cYun Huanga,cYan Zhangb,c( )Mingming Gaod( )Pin Lüa( )
Postdoctoral Station for Basic Medicine, Cardiovascular Medical Science Center, Department of Cell Biology, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
Hebei Key Laboratory of Forensic Medicine, College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, China
Hebei Food Safety Key Laboratory, Hebei Food Inspection and Research Institute, Shijiazhuang 050091, China
Laboratory of Lipid Metabolism, Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Shijiazhuang 050017, China

# These authors have contributed equally to this work.

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Abstract

Intermittent fasting can benefit breast cancer patients undergoing chemotherapy or immunotherapy. However, it is still uncertain how to select immunotherapy drugs to combine with intermittent fasting. Herein we observed that two cycles of fasting treatment significantly inhibited breast tumor growth and lung tissue metastasis, as well as prolonged overall survival in mice bearing 4T1 and 4T07 breast cancer. During this process, both the immunosuppressive monocytic- (M-) and granulocytic- (G-) myeloid-derived suppressor cell (MDSC) decreased, accompanied by an increase in interleukin (IL) 7R+ and granzyme B+ T cells in the tumor microenvironment. Interestingly, we observed that Ly6Glow G-MDSC sharply decreased after fasting treatment, and the cell surface markers and protein mass spectrometry data showed potential therapeutic targets. Mechanistic investigation revealed that glucose metabolism restriction suppressed the splenic granulocyte-monocyte progenitor and the generation of colony-stimulating factors and IL-6, which both contributed to the accumulation of G-MDSC. On the other hand, glucose metabolism restriction can directly induce the apoptosis of Ly6Glow G-MDSC, but not Ly6Ghigh subsets. In summary, these results suggest that glucose metabolism restriction induced by fasting treatment attenuates the immune-suppressive milieu and enhances the activation of CD3+ T cells, providing potential solutions for enhancing immune-based cancer interventions.

Electronic Supplementary Material

Download File(s)
fshw-13-4-2327_ESM2.pdf (86.5 KB)

References

[1]

M. Tajan, K.H. Vousden, Dietary approaches to cancer therapy, Cancer Cell 37(6) (2020) 767-785. https://doi.org/10.1016/j.ccell.2020.04.005.

[2]

U. Kämmerer, R.J. Klement, F.T. Joos, et al., Low carb and ketogenic diets increase quality of life, physical performance, body composition, and metabolic health of women with breast cancer, Nutrients 13(3) (2021) 1029. https://doi.org/10.3390/nu13031029.

[3]

T. Komatsu, S. Park, H. Hayashi, et al., Mechanisms of calorie restriction: a review of genes required for the life-extending and tumor-inhibiting effects of calorie restriction, Nutrients 11(12) (2019) 3068. https://doi.org/10.3390/nu11123068.

[4]

M. Wei, S. Brandhorst, M. Shelehchi, et al., Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease, Sci. Transl. Med. 15 (2017) eaai8700. https://doi.org/10.1126/scitranslmed.aai8700.

[5]

A. Nencioni, I. Caffa, S. Cortellino, et al., Fasting and cancer: molecular mechanisms and clinical application, Nat. Rev. Cancer 18(11) (2018) 707-719. https://doi.org/10.1038/s41568-018-0061-0.

[6]

M.P. Mattson, V.D. Longo, M. Harvie. Impact of intermittent fasting on health and disease processes. Ageing Res. Rev. 39 (2017) 46-58. https://doi.org/10.1016/j.arr.2016.10.005.

[7]

R.D. Cabo, M.P. Mattson. Effects of intermittent fasting on health, aging, and disease, N. Engl. J. Med. 381(26) (2019) 2541-2551. https://doi.org/10.1056/NEJMra1905136.

[8]

C. Lee, L. Raffaghello, S. Brandhorst et al., Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy, Sci. Transl. Med. 4(124) (2012) 124ra27. https://doi.org/10.1126/scitranslmed.3003293.

[9]

V.D. Longo, C.E. Finch. Evolutionary medicine: from dwarf model systems to healthy centenarians?, Science 299(5611) (2003) 1342-1346. https://doi.org/10.1126/science.1077991.

[10]

V.D. Longo, M.P. Mattson. Fasting: molecular mechanisms and clinical applications, Cell Metab. 19(2) (2014) 181-192. https://doi.org/10.1016/j.cmet.2013.12.008.

[11]

D.R. Wang, X.L. Wu, Y.L. Sun. Therapeutic targets and biomarkers of tumor immunotherapy: response versus non-response, Signal Transduct. Target Ther. 7(1) (2022) 331. https://doi.org/10.1038/s41392-022-01136-2.

[12]

D.S. Chen, I. Mellman. Oncology meets immunology: the cancer-immunity cycle, Immunity 39(1) (2013) 1-10. https://doi.org/10.1016/j.immuni.2013.07.012.

[13]

C.V. Rothlin, S Ghosh, Lifting the innate immune barriers to antitumor immunity, J. Immunother Cancer 8(1) (2020) e000695. https://doi.org/10.1136/jitc-2020-000695.

[14]

H.G. Wiktorin, M.S. Nilsson, R Kiffin, et al., Histamine targets myeloid-derived suppressor cells and improves the anti-tumor efficacy of PD-1/PD-L1 checkpoint blockade, Cancer Immunol. Immunother 68(2) (2019) 163-174. https://doi.org/10.1007/s00262-018-2253-6.

[15]

R.J. Davis, E.C. Moore, P.E. Clavijo, et al., Anti-PD-L1 efficacy can be enhanced by inhibition of myeloid-derived suppressor cells with a selective inhibitor of PI3Kδ/γ, Cancer Res, 77(10) (2017) 2607-2619. https://doi.org/10.1158/0008-5472.can-16-2534.

[16]

C. Fu, Z. Fu, C. Jiang, et al., CD205+ polymorphonuclear myeloid-derived suppressor cells suppress antitumor immunity by overexpressing GLUT3, Cancer Sci. 112(3) (2021) 1011-1025. https://doi.org/10.1111/cas.14783.

[17]

V. Bronte, S. Brandau, S.H. Chen, et al., Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards, Nat. Commun. 6/7 (2016) 12150. https://doi.org/10.1038/ncomms12150.

[18]

M. Evrard, I.W.H. Kwok, S.Z. Chong, et al., Developmental analysis of bone marrow neutrophils reveals populations specialized in expansion, trafficking, and effector functions, Immunity 48(2) (2018) 364-379.e8. https://doi.org/10.1016/j.immuni.2018.02.002.

[19]

J. Zhou, Y. Nefedova, A. Lei, et al., Neutrophils and PMN-MDSC: their biological role and interaction with stromal cells, Semin. Immunol. 35 (2018) 19-28. https://doi.org/10.1016/j.smim.2017.12.004.

[20]

J. Cai J, D. Wang, G. Zhang, et al., The role of PD-1/PD-L1 axis in Treg development and function: implications for cancer immunotherapy, Onco. Targets Ther. 12 (2019) 8437-8445. https://doi.org/10.2147/ott.s221340.

[21]

S. Han, A. Toker, Z.Q. Liu, et al., Turning the tide against regulatory T cells, Front. Oncol. 9 (2019) 279. https://doi.org/10.3389/fonc.2019.00279.

[22]

T.L. Whiteside. FOXP3+ Treg as a therapeutic target for promoting anti-tumor immunity, Expert. Opin. Ther. Targets 22(4) (2018) 353-363. https://doi.org/10.1080/14728222.2018.1451514.

[23]

A.D. Waldman, J.M. Fritz, M.J. Lenardo. A guide to cancer immunotherapy: from T cell basic science to clinical practice, Nat. Rev. Immunol. 20(11) (2020) 651-668. https://doi.org/10.1038/s41577-020-0306-5.

[24]

W.J. Turbitt, Y. Xu, D.M. Sosnoski, et al., Physical activity plus energy restriction prevents 4T1.2 mammary tumor progression, MDSC accumulation, and an immunosuppressive tumor microenvironment, Cancer Prev. Res. (Phila) 12(8) (2019) 493-506. https://doi.org/10.1158/1940-6207.capr-17-0233.

[25]

C.H. Guo, S. Hsia, C.H. Chung, et al., Nutritional supplements in combination with chemotherapy or targeted therapy reduces tumor progression in mice bearing triple-negative breast cancer, J. Nutr. Biochem. 87 (2021) 108504. https://doi.org/10.1016/j.jnutbio.2020.108504.

[26]

M.J. Welters, T.C.V.D. Sluis, H.V. Meir, et al., Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses, Sci. Transl. Med. 8(334) (2016) 334ra52. https://doi.org/10.1126/scitranslmed.aad8307.

[27]

C.W. Cheng, G.B. Adams, L. Perin, et al., Prolonged fasting reduces IGF-1/PKA to promote hematopoietic-stem-cell-based regeneration and reverse immunosuppression, Cell Stem Cell 14(6) (2014) 810-823. https://doi.org/10.1016/j.stem.2014.04.014.

[28]

F. Pietrocola, J. Pol, E. Vacchelli, et al., Caloric restriction mimetics enhance anticancer immunosurveillance, Cancer Cell 30(1) (2016) 147-160. https://doi.org/10.1016/j.ccell.2016.05.016.

[29]

J.M. Englert, J.D. Powell. Hunger pains: stimulating the appetite of the immune system for cancer, Cancer Cell 30(1) (2016) 13-15. https://doi.org/10.1016/j.ccell.2016.06.019.

[30]

S.D Groot, H. Pijl, J.J.M.V.D. Hoeven, et al., Effects of short-term fasting on cancer treatment. J Exp Clin Cancer Res 38(1) (2019) 209. https://doi.org/10.1186/s13046-019-1189-9.

[31]

S.D. Biase, C. Lee, S. Brandhorst, et al., Fasting-mimicking diet reduces HO-1 to promote T Cell-mediated tumor cytotoxicity, Cancer Cell 30(1) (2016) 136-146. https://doi.org/10.1016/j.ccell.2016.06.005.

[32]

D. Guo, Y. Tong, X. Jiang, et al., Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IκBα, Cell Metab. 34(9) (2022) 1312-1324.e6. https://doi.org/10.1016/j.cmet.2022.08.002.

[33]

B.I. Reinfeld, M.Z. Madden, M.M. Wolf, et al., Cell-programmed nutrient partitioning in the tumour microenvironment, Nature 593 (2021) 282-288. https://doi.org/10.1038/s41586-021-03442-1.

[34]

D.M. Euhus, C. Hudd, M.C. Laregina, et al., Tumor measurement in the nude mouse, J. Surg. Oncol. 31(4) (1986) 229-234. https://doi.org/10.1002/jso.2930310402.

[35]

D. Sheng, W. Ma, R. Zhang, et al., Ccl3 enhances docetaxel chemosensitivity in breast cancer by triggering proinflammatory macrophage polarization, J. Immunother. Cancer 10(5) (2022) e003793. https://doi.org/10.1136/jitc-2021-003793.

[36]

C.R. Millrud, M. Mehmeti, K. Leandersson. Docetaxel promotes the generation of anti-tumorigenic human macrophages. Exp. Cell Res. 362(2) (2018) 525-531. https://doi.org/10.1016/j.yexcr.2017.12.018.

[37]

D. Alizadeh D, M. Trad, N.T. Hanke, et al., Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer, Cancer Res. 74(1) (2014) 104-118. https://doi.org/10.1158/0008-5472.can-13-1545.

[38]

K. Zheng K, Q. Li, D. Lin, et al., Peptidomic analysis of pilose antler and its inhibitory effect on triple-negative breast cancer at multiple sites, Food Funct. 11(9) (2020) 7481-7494. https://doi.org/10.1039/d0fo01531h.

[39]

Q. Liang, Q. Zhao, X. Hao, et al., The effect of flammulina velutipes polysaccharide on immunization analyzed by intestinal flora and proteomics, Front. Nutr. 28 (2022) 841230. https://doi.org/10.3389/fnut.2022.841230.

[40]

J. Steenbrugge, E.A.D. Jaeghere, E. Meyer, et al., Splenic hematopoietic and stromal cells in cancer progression, Cancer Res. 81(1) (2021) 27-34. https://doi.org/10.1158/0008-5472.can-20-2339.

[41]

C. Wu, H.H. Ning, M.Y. Liu, et al., Spleen mediates a distinct hematopoietic progenitor response supporting tumor-promoting myelopoiesis, J. Clin. Invest. 128(8) (2018) 3425-3438. https://doi.org/10.1172/jci97973.

[42]

Z. Zou, T. Tao, H. Li, et al., mTOR signaling pathway and mTOR inhibitors in cancer: progress and challenges, Cell Biosci. 10 (2020) 31. https://doi.org/10.1186/s13578-020-00396-1.

[43]

G.Y. Liu, D.M. Sabatini. mTOR at the nexus of nutrition, growth, ageing and disease, Nat. Rev. Mol. Cell. Biol. 21(4) (2020) 183-203. https://doi.org/10.1038/s41580-019-0199-y.

[44]

M.S.D. Lorenzo, E. Baljinnyam, D.E. Vatner, et al., Caloric restriction reduces growth of mammary tumors and metastases, Carcinogenesis 32(9) (2011) 1381-1387. https://doi.org/10.1093/carcin/bgr107.

[45]

R.J. Colman, R.M. Anderson, S.C. Johnson, et al., Caloric restriction delays disease onset and mortality in rhesus monkeys, Science 325(5937) (2009) 201-204. https://doi.org/10.1126/science.1173635.

[46]

C. Lee C, V.D. Longo. Fasting vs dietary restriction in cellular protection and cancer treatment: from model organisms to patients, Oncogene 30(30) (2011) 3305-3316. https://doi.org/10.1038/onc.2011.91.

[47]

J. Song, S.F. Ke, C.C. Zhou, et al., Nicotinamide phosphoribosyltransferase is required for the calorie restriction-mediated improvements in oxidative stress, mitochondrial biogenesis, and metabolic adaptation, J. Gerontol. A Biol. Sci. Med. Sci. 69(1) (2014) 44-57. https://doi.org/10.1093/gerona/glt122.

[48]

S. Brandhorst, I.Y. Choi, M. Wei, et al., A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell. Metab. 22(1) (2015) 86-99. https://doi.org/10.1016/j.cmet.2015.05.012.

[49]

X. Zhao, J. Yang, R. Huang, et al., The role and its mechanism of intermittent fasting in tumors: friend or foe? Cancer Biol. Med. 18(1) (2021) 63-73. https://doi.org/10.20892/j.issn.2095-3941.2020.0250.

[50]

H.A. Watson, R.R.P. Durairaj, J. Ohme, et al., L-selectin enhanced T cells improve the efficacy of cancer immunotherapy, Front. Immunol. 12 (2019) 1321. https://doi.org/10.3389/fimmu.2019.01321.

[51]

M. Ohara, Y. Yamaguchi, K. Matsuura, et al., Possible involvement of regulatory T cells in tumor onset and progression in primary breast cancer, Cancer Immunol. Immunother 58(3) (2009) 441-447. https://doi.org/10.1007/s00262-008-0570-x.

[52]

V. Hashemi, L.A. Maleki, M. Esmaily, et al., Regulatory T cells in breast cancer as a potent anti-cancer therapeutic target, Int. Immunopharmacol. 78 (2020) 106087. https://doi.org/10.1016/j.intimp.2019.106087.

[53]

W. Liu, A.L. Putnam, X.Y. Zhou, et al., CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells, J. Exp. Med. 203(7) (2006) 1701-1711. https://doi.org/10.1084/jem.20060772.

[54]

S.G. Kalathil, A.A. Lugade, A. Miller, et al., PD-1+ and Foxp3+ T cell reduction correlates with survival of HCC patients after sorafenib therapy, JCI Insight 1(11) (2016) https://doi.org/10.1172/jci.insight.86182.

[55]

M.G.V. Heiden, J.W. Locasale, K.D. Swanson, et al., Evidence for an alternative glycolytic pathway in rapidly proliferating cells, Science 329 (2010) 1492-1499. https://doi.org/10.1126/science.1188015.

[56]

R.G. Jones, C.B. Thompson. Tumor suppressors and cell metabolism: a recipe for cancer growth, Genes Dev. 23(5) (2009) 537-548. https://doi.org/10.1101/gad.1756509.

[57]

W. Li, T. Tanikawa, I. Kryczek, et al., Aerobic glycolysis controls myeloid-derived suppressor cells and tumor immunity via a specific CEBPB isoform in triple-negative breast cancer, Cell Metab. 28(1) (2018) 87-103.e6. https://doi.org/10.1016/j.cmet.2018.04.022.

[58]

S.L. Jian, W.W. Chen, Y.C. Su, et al., Glycolysis regulates the expansion of myeloid-derived suppressor cells in tumor-bearing hosts through prevention of ROS-mediated apoptosis, Cell Death Dis. 8(5) (2017) e2779. https://doi.org/10.1038/cddis.2017.192.

[59]

N. Patsoukis, K. Bardhan, P. Chatterjee, et al., PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation, Nat. Commun. 26 (2015) 6692. https://doi.org/10.1038/ncomms7692.

[60]

X. Yu, X.L. Teng, F. Wang, et al., Metabolic control of regulatory T cell stability and function by TRAF3IP3 at the lysosome, J. Exp. Med. 215(9) (2018) 2463-2476. https://doi.org/10.1084/jem.20180397.

[61]

S.D. Saibil, P.S. Ohashi. Targeting T cell activation in immuno-oncology, Curr. Oncol. 27(Suppl 2) (2020) S98-S105. https://doi.org/10.3747/co.27.5285.

[62]

J. Fucikova, O. Kepp, L. Kasikova, et al., Detection of immunogenic cell death and its relevance for cancer therapy, Cell Death Dis. 11(11) (2020) 1013. https://doi.org/10.1038/s41419-020-03221-2.

[63]

N.D. Gruil, H. Pijl, S.H.V.D. Burg, et al., Short-term fasting synergizes with solid cancer therapy by boosting antitumor immunity, Cancers 14(6) (2022) 1390. https://doi.org/10.3390/cancers14061390.

Food Science and Human Wellness
Pages 2327-2345
Cite this article:
Fu C, Liang Z, Niu Z, et al. Intermittent fasting boosts antitumor immunity by restricting CD11b+Ly6Clow Ly6Glow cell viability through glucose metabolism in murine breast tumor model. Food Science and Human Wellness, 2024, 13(4): 2327-2345. https://doi.org/10.26599/FSHW.2022.9250194

722

Views

76

Downloads

0

Crossref

0

Web of Science

0

Scopus

0

CSCD

Altmetrics

Received: 29 April 2023
Revised: 28 May 2023
Accepted: 17 June 2023
Published: 20 May 2024
© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return