Skip to main content
Top
Published in: Discover Oncology 1/2023

Open Access 01-12-2023 | Breast Cancer | Research

GPR81-mediated reprogramming of glucose metabolism contributes to the immune landscape in breast cancer

Authors: Xiaofeng li, Yiwen Chen, Ting Wang, Zifan Liu, Guotao Yin, Ziyang Wang, Chunxiao Sui, Lei Zhu, Wei Chen

Published in: Discover Oncology | Issue 1/2023

Login to get access

Abstract

Background

Local tumor microenvironment (TME) plays a crucial role in immunotherapy for breast cancer (BC). Whereas, the molecular mechanism responsible for the crosstalk between BC cells and surrounding immune cells remains unclear. The present study aimed to determine the interplay between GPR81-mediated glucometabolic reprogramming of BC and the immune landscape in TME.

Materials and Methods

Immunohistochemistry (IHC) assay was first performed to evaluate the association between GPR81 and the immune landscape. Then, several stable BC cell lines with down-regulated GPR81 expression were established to directly identify the role of GPR81 in glucometabolic reprogramming, and western blotting assay was used to detect the underlying molecular mechanism. Finally, a transwell co-culture system confirmed the crosstalk between glucometabolic regulation mediated by GPR81 in BC and induced immune attenuation.

Results

IHC analysis demonstrated that the representation of infiltrating CD8+ T cells and FOXP3+ T cells were dramatically higher in BC with a triple negative (TN) subtype in comparison with that with a non-TN subtype (P < 0.001). Additionally, the ratio of infiltrating CD8+ to FOXP3+ T cells was significantly negatively associated with GPR81 expression in BC with a TN subtype (P < 0.001). Furthermore, GPR81 was found to be substantially correlated with the glycolytic capability (P < 0.001) of BC cells depending on a Hippo-YAP signaling pathway (P < 0.001). In the transwell co-culture system, GPR81-mediated reprogramming of glucose metabolism in BC significantly contributed to a decreased proportion of CD8+ T (P < 0.001) and an increased percentage of FOXP3+ T (P < 0.001) in the co-cultured lymphocytes.

Conclusion

Glucometabolic reprogramming through a GPR81-mediated Hippo-YAP signaling pathway was responsible for the distinct immune landscape in BC. GPR81 was a potential biomarker to stratify patients before immunotherapy to improve BC’s clinical prospect.
Appendix
Available only for authorised users
Literature
1.
go back to reference Siegel RL, Miller KD, Fuchs HE. Jemal A (2021) cancer statistics. CA Cancer J Clin. 2021;71(1):7–33.PubMedCrossRef Siegel RL, Miller KD, Fuchs HE. Jemal A (2021) cancer statistics. CA Cancer J Clin. 2021;71(1):7–33.PubMedCrossRef
2.
go back to reference Sun N, Gao P, Li Y, et al. Screening and identification of key common and specific genes and their prognostic roles in different molecular subtypes of breast cancer. Front Mol Biosci. 2021;8: 619110.PubMedPubMedCentralCrossRef Sun N, Gao P, Li Y, et al. Screening and identification of key common and specific genes and their prognostic roles in different molecular subtypes of breast cancer. Front Mol Biosci. 2021;8: 619110.PubMedPubMedCentralCrossRef
3.
go back to reference Fumagalli C, Ranghiero A, Gandini S, et al. Inter-tumor genomic heterogeneity of breast cancers: comprehensive genomic profile of primary early breast cancers and relapses. Breast Cancer Res. 2020;22(1):107.PubMedPubMedCentralCrossRef Fumagalli C, Ranghiero A, Gandini S, et al. Inter-tumor genomic heterogeneity of breast cancers: comprehensive genomic profile of primary early breast cancers and relapses. Breast Cancer Res. 2020;22(1):107.PubMedPubMedCentralCrossRef
4.
go back to reference Prieto-Vila M, Usuba W, Takahashi RU, et al. Single-cell analysis reveals a preexisting drug-resistant subpopulation in the luminal breast cancer subtype. Cancer Res. 2019;79(17):4412–25.PubMedCrossRef Prieto-Vila M, Usuba W, Takahashi RU, et al. Single-cell analysis reveals a preexisting drug-resistant subpopulation in the luminal breast cancer subtype. Cancer Res. 2019;79(17):4412–25.PubMedCrossRef
5.
go back to reference Masoumi E, Tahaghoghi-Hajghorbani S, Jafarzadeh L, Sanaei MJ, Pourbagheri-Sigaroodi A, Bashash D. The application of immune checkpoint blockade in breast cancer and the emerging role of nanoparticle. J Control Release. 2021;340:168–87.PubMedCrossRef Masoumi E, Tahaghoghi-Hajghorbani S, Jafarzadeh L, Sanaei MJ, Pourbagheri-Sigaroodi A, Bashash D. The application of immune checkpoint blockade in breast cancer and the emerging role of nanoparticle. J Control Release. 2021;340:168–87.PubMedCrossRef
6.
go back to reference Pernas S, Tolaney SM. Clinical trial data and emerging strategies: HER2-positive breast cancer. Breast Cancer Res Treat. 2022;193(2):281–91.PubMedCrossRef Pernas S, Tolaney SM. Clinical trial data and emerging strategies: HER2-positive breast cancer. Breast Cancer Res Treat. 2022;193(2):281–91.PubMedCrossRef
7.
go back to reference Wise J. NICE allows immunotherapy treatment for small group of patients with advanced triple negative breast cancer. BMJ. 2022;377: o1311.PubMedCrossRef Wise J. NICE allows immunotherapy treatment for small group of patients with advanced triple negative breast cancer. BMJ. 2022;377: o1311.PubMedCrossRef
8.
go back to reference Yang A, Wu M, Ni M, et al. A risk scoring system based on tumor microenvironment cells to predict prognosis and immune activity in triple-negative breast cancer. Breast Cancer. 2022;29(3):468–77.PubMedCrossRef Yang A, Wu M, Ni M, et al. A risk scoring system based on tumor microenvironment cells to predict prognosis and immune activity in triple-negative breast cancer. Breast Cancer. 2022;29(3):468–77.PubMedCrossRef
9.
go back to reference Huang K, Hu M, Chen J, et al. Multi-omics perspective reveals the different patterns of tumor immune microenvironment based on programmed death ligand 1 (PD-L1) expression and predictor of responses to immune checkpoint blockade across pan-cancer. Int J Mol Sci. 2021;22(10):5158.PubMedPubMedCentralCrossRef Huang K, Hu M, Chen J, et al. Multi-omics perspective reveals the different patterns of tumor immune microenvironment based on programmed death ligand 1 (PD-L1) expression and predictor of responses to immune checkpoint blockade across pan-cancer. Int J Mol Sci. 2021;22(10):5158.PubMedPubMedCentralCrossRef
10.
11.
go back to reference Zhao J, Meisel J, Guo Y, et al. Evaluation of PD-L1, tumor-infiltrating lymphocytes, and CD8+ and FOXP3+ immune cells in HER2-positive breast cancer treated with neoadjuvant therapies. Breast Cancer Res Treat. 2020;183(3):599–606.PubMedCrossRef Zhao J, Meisel J, Guo Y, et al. Evaluation of PD-L1, tumor-infiltrating lymphocytes, and CD8+ and FOXP3+ immune cells in HER2-positive breast cancer treated with neoadjuvant therapies. Breast Cancer Res Treat. 2020;183(3):599–606.PubMedCrossRef
12.
go back to reference Criscitiello C, Vingiani A, Maisonneuve P, Viale G, Viale G, Curigliano G. Tumor-infiltrating lymphocytes (TILs) in ER+/HER2- breast cancer. Breast Cancer Res Treat. 2020;183(2):347–54.PubMedCrossRef Criscitiello C, Vingiani A, Maisonneuve P, Viale G, Viale G, Curigliano G. Tumor-infiltrating lymphocytes (TILs) in ER+/HER2- breast cancer. Breast Cancer Res Treat. 2020;183(2):347–54.PubMedCrossRef
13.
go back to reference Baldominos P, Barbera-Mourelle A, Barreiro O, et al. Quiescent cancer cells resist T cell attack by forming an immunosuppressive niche. Cell. 2022;185(10):1694-1708.e19.PubMedCrossRef Baldominos P, Barbera-Mourelle A, Barreiro O, et al. Quiescent cancer cells resist T cell attack by forming an immunosuppressive niche. Cell. 2022;185(10):1694-1708.e19.PubMedCrossRef
14.
go back to reference Wei F, Wang D, Wei J, et al. Metabolic crosstalk in the tumor microenvironment regulates antitumor immunosuppression and immunotherapy resisitance. Cell Mol Life. 2021;78(1):173–93.CrossRef Wei F, Wang D, Wei J, et al. Metabolic crosstalk in the tumor microenvironment regulates antitumor immunosuppression and immunotherapy resisitance. Cell Mol Life. 2021;78(1):173–93.CrossRef
15.
go back to reference Morrissey SM, Zhang F, Ding C, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021;33(10):2040–58.PubMedPubMedCentralCrossRef Morrissey SM, Zhang F, Ding C, et al. Tumor-derived exosomes drive immunosuppressive macrophages in a pre-metastatic niche through glycolytic dominant metabolic reprogramming. Cell Metab. 2021;33(10):2040–58.PubMedPubMedCentralCrossRef
16.
go back to reference Corchado-Cobos R, García-Sancha N, Mendiburu-Eliçabe M, et al. Pathophysiological integration of metabolic reprogramming in breast cancer. Cancers (Basel). 2022;14(2):322.PubMedCrossRef Corchado-Cobos R, García-Sancha N, Mendiburu-Eliçabe M, et al. Pathophysiological integration of metabolic reprogramming in breast cancer. Cancers (Basel). 2022;14(2):322.PubMedCrossRef
17.
go back to reference Wu Z, Wu J, Zhao Q, Fu S, Jin J. Emerging roles of aerobic glycolysis in breast cancer. Clin Transl Oncol. 2020;22(5):631–46.PubMedCrossRef Wu Z, Wu J, Zhao Q, Fu S, Jin J. Emerging roles of aerobic glycolysis in breast cancer. Clin Transl Oncol. 2020;22(5):631–46.PubMedCrossRef
18.
go back to reference Naik A, Decock J. Lactate metabolism and immune modulation in breast cancer: a focused review on triple negative breast tumors. Front Oncol. 2020;10: 598626.PubMedPubMedCentralCrossRef Naik A, Decock J. Lactate metabolism and immune modulation in breast cancer: a focused review on triple negative breast tumors. Front Oncol. 2020;10: 598626.PubMedPubMedCentralCrossRef
19.
go back to reference Hayes C, Donohoe CL, Davern M, Donlon NE. The oncogenic and clinical implications of lactate induced immunosuppression in the tumour microenvironment. Cancer Lett. 2021;500:75–86.PubMedCrossRef Hayes C, Donohoe CL, Davern M, Donlon NE. The oncogenic and clinical implications of lactate induced immunosuppression in the tumour microenvironment. Cancer Lett. 2021;500:75–86.PubMedCrossRef
20.
21.
22.
go back to reference Feng J, Yang H, Zhang Y, et al. Tumor cell-derived lactate induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer cells. Oncogene. 2017;36(42):5829–39.PubMedCrossRef Feng J, Yang H, Zhang Y, et al. Tumor cell-derived lactate induces TAZ-dependent upregulation of PD-L1 through GPR81 in human lung cancer cells. Oncogene. 2017;36(42):5829–39.PubMedCrossRef
23.
go back to reference Yang X, Lu Y, Hang J, et al. Lactate-modulated immunosuppression of myeloid-derived suppressor cells contributes to the radioresistance of pancreatic cancer. Cancer Immunol Res. 2020;8(11):1440–51.PubMedCrossRef Yang X, Lu Y, Hang J, et al. Lactate-modulated immunosuppression of myeloid-derived suppressor cells contributes to the radioresistance of pancreatic cancer. Cancer Immunol Res. 2020;8(11):1440–51.PubMedCrossRef
24.
go back to reference Longhitano L, Forte S, Orlando L, et al. The crosstalk between GPR81/IGFBP6 promotes breast cancer progression by modulating lactate metabolism and oxidative stress. Antioxidants. 2022;11(2):275.PubMedPubMedCentralCrossRef Longhitano L, Forte S, Orlando L, et al. The crosstalk between GPR81/IGFBP6 promotes breast cancer progression by modulating lactate metabolism and oxidative stress. Antioxidants. 2022;11(2):275.PubMedPubMedCentralCrossRef
25.
go back to reference Brown TP, Bhattacharjee P, Ramachandran S, et al. The lactate receptor GPR81 promotes breast cancer growth via a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment. Oncogene. 2020;39(16):3292–304.PubMedCrossRef Brown TP, Bhattacharjee P, Ramachandran S, et al. The lactate receptor GPR81 promotes breast cancer growth via a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment. Oncogene. 2020;39(16):3292–304.PubMedCrossRef
26.
27.
go back to reference Serrano-Carbajal EA, Espinal-Enríquez J, Hernández-Lemus E. Targeting metabolic deregulation landscapes in breast cancer subtypes. Front Oncol. 2020;10:97.PubMedPubMedCentralCrossRef Serrano-Carbajal EA, Espinal-Enríquez J, Hernández-Lemus E. Targeting metabolic deregulation landscapes in breast cancer subtypes. Front Oncol. 2020;10:97.PubMedPubMedCentralCrossRef
29.
30.
go back to reference Cappelletti V, Iorio E, Miodini P, Silvestri M, Dugo M, Daidone MG. Metabolic footprints and molecular subtypes in breast cancer. Dis Markers. 2017;2017:7687851.CrossRefPubMedPubMedCentral Cappelletti V, Iorio E, Miodini P, Silvestri M, Dugo M, Daidone MG. Metabolic footprints and molecular subtypes in breast cancer. Dis Markers. 2017;2017:7687851.CrossRefPubMedPubMedCentral
31.
go back to reference Adams TA, Vail PJ, Ruiz A, et al. Composite analysis of immunological and metabolic markers defines novel subtypes of triple negative breast cancer. Mod Pathol. 2018;31(2):288–98.PubMedCrossRef Adams TA, Vail PJ, Ruiz A, et al. Composite analysis of immunological and metabolic markers defines novel subtypes of triple negative breast cancer. Mod Pathol. 2018;31(2):288–98.PubMedCrossRef
32.
go back to reference Lee YJ, Shin KJ, Park SA, et al. G-protein-coupled receptor 81 promotes a malignant phenotype in breast cancer through angiogenic factor secretion. Oncotarget. 2016;7(43):70898–911.PubMedPubMedCentralCrossRef Lee YJ, Shin KJ, Park SA, et al. G-protein-coupled receptor 81 promotes a malignant phenotype in breast cancer through angiogenic factor secretion. Oncotarget. 2016;7(43):70898–911.PubMedPubMedCentralCrossRef
33.
35.
36.
go back to reference Luen SJ, Savas P, Fox SB, Salgado R, Loi S. Tumour-infiltrating lymphocytes and the emerging role of immunotherapy in breast cancer. Pathology. 2017;49(2):141–55.PubMedCrossRef Luen SJ, Savas P, Fox SB, Salgado R, Loi S. Tumour-infiltrating lymphocytes and the emerging role of immunotherapy in breast cancer. Pathology. 2017;49(2):141–55.PubMedCrossRef
37.
go back to reference Lu Z, Xu S, Shao W, et al. Deep-learning-based characterization of tumor-infiltrating lymphocytes in breast cancers from histopathology images and multiomics data. JCO Clin Cancer Inform. 2020;4:480–90.PubMedCrossRef Lu Z, Xu S, Shao W, et al. Deep-learning-based characterization of tumor-infiltrating lymphocytes in breast cancers from histopathology images and multiomics data. JCO Clin Cancer Inform. 2020;4:480–90.PubMedCrossRef
38.
go back to reference Morrot A, da Fonseca LM, Salustiano EJ, et al. Metabolic symbiosis and immunomodulation: how tumor cell-derived lactate may disturb innate and adaptive immune responses. Front Oncol. 2018;8:81.PubMedPubMedCentralCrossRef Morrot A, da Fonseca LM, Salustiano EJ, et al. Metabolic symbiosis and immunomodulation: how tumor cell-derived lactate may disturb innate and adaptive immune responses. Front Oncol. 2018;8:81.PubMedPubMedCentralCrossRef
39.
go back to reference Baxevanis CN, Fortis SP, Perez SA. The balance between breast cancer and the immune system: challenges for prognosis and clinical benefit from immunotherapies. Semin Cancer Biol. 2021;72:76–89.PubMedCrossRef Baxevanis CN, Fortis SP, Perez SA. The balance between breast cancer and the immune system: challenges for prognosis and clinical benefit from immunotherapies. Semin Cancer Biol. 2021;72:76–89.PubMedCrossRef
40.
go back to reference Švajger U, Tešić N, Rožman P. Programmed death ligand 1 (PD-L1) plays a vital part in DC tolerogenicity induced by IFN-γ. Int Immunopharmacol. 2021;99: 107978.PubMedCrossRef Švajger U, Tešić N, Rožman P. Programmed death ligand 1 (PD-L1) plays a vital part in DC tolerogenicity induced by IFN-γ. Int Immunopharmacol. 2021;99: 107978.PubMedCrossRef
41.
go back to reference Yang J, Chen J, Liang H, Yu Y. Nasopharyngeal cancer cell-derived exosomal PD-L1 inhibits CD8+ T cell activity and promotes immune escape. Cancer Sci. 2022;113(9):3044–54.PubMedPubMedCentralCrossRef Yang J, Chen J, Liang H, Yu Y. Nasopharyngeal cancer cell-derived exosomal PD-L1 inhibits CD8+ T cell activity and promotes immune escape. Cancer Sci. 2022;113(9):3044–54.PubMedPubMedCentralCrossRef
42.
go back to reference Turbitt WJ, Demark-Wahnefried W, Peterson CM, Norian LA. Targeting glucose metabolism to enhance immunotherapy: emerging evidence on intermittent fasting and calorie restriction mimetics. Front Immunol. 2019;10:1402.PubMedPubMedCentralCrossRef Turbitt WJ, Demark-Wahnefried W, Peterson CM, Norian LA. Targeting glucose metabolism to enhance immunotherapy: emerging evidence on intermittent fasting and calorie restriction mimetics. Front Immunol. 2019;10:1402.PubMedPubMedCentralCrossRef
43.
go back to reference Renner K, Bruss C, Schnell A, et al. Restricting glycolysis preserves t cell effector functions and augments checkpoint therapy. Cell Rep. 2019;29(1):135-50.e9.PubMedCrossRef Renner K, Bruss C, Schnell A, et al. Restricting glycolysis preserves t cell effector functions and augments checkpoint therapy. Cell Rep. 2019;29(1):135-50.e9.PubMedCrossRef
44.
go back to reference Lundø K, Trauelsen M, Pedersen SF, Schwartz TW. Why warburg works: lactate controls immune evasion through GPR81. Cell Metab. 2020;31(4):666–8.PubMedCrossRef Lundø K, Trauelsen M, Pedersen SF, Schwartz TW. Why warburg works: lactate controls immune evasion through GPR81. Cell Metab. 2020;31(4):666–8.PubMedCrossRef
45.
go back to reference Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162(6):1229–41.PubMedPubMedCentralCrossRef Chang CH, Qiu J, O’Sullivan D, Buck MD, Noguchi T, Curtis JD, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162(6):1229–41.PubMedPubMedCentralCrossRef
Metadata
Title
GPR81-mediated reprogramming of glucose metabolism contributes to the immune landscape in breast cancer
Authors
Xiaofeng li
Yiwen Chen
Ting Wang
Zifan Liu
Guotao Yin
Ziyang Wang
Chunxiao Sui
Lei Zhu
Wei Chen
Publication date
01-12-2023
Publisher
Springer US
Published in
Discover Oncology / Issue 1/2023
Print ISSN: 1868-8497
Electronic ISSN: 2730-6011
DOI
https://doi.org/10.1007/s12672-023-00709-z

Other articles of this Issue 1/2023

Discover Oncology 1/2023 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine