Cargando…

Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment

BACKGROUND: A close metabolic interaction between cancer and immune cells in the tumor microenvironment (TME) plays a pivotal role in cancer immunity. Herein, we have comprehensively investigated the glucose metabolic features of the TME at the single-cell level to discover feasible metabolic target...

Descripción completa

Detalles Bibliográficos
Autores principales: Choi, Hongyoon, Na, Kwon Joong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8710507/
https://www.ncbi.nlm.nih.gov/pubmed/34966676
http://dx.doi.org/10.3389/fonc.2021.769393
_version_ 1784623169882030080
author Choi, Hongyoon
Na, Kwon Joong
author_facet Choi, Hongyoon
Na, Kwon Joong
author_sort Choi, Hongyoon
collection PubMed
description BACKGROUND: A close metabolic interaction between cancer and immune cells in the tumor microenvironment (TME) plays a pivotal role in cancer immunity. Herein, we have comprehensively investigated the glucose metabolic features of the TME at the single-cell level to discover feasible metabolic targets for the tumor immune status. METHODS: We examined expression levels of glucose transporters (GLUTs) in various cancer types using The Cancer Genome Atlas (TCGA) data and single-cell RNA-seq (scRNA-seq) datasets of human cancer tissues including melanoma, head and neck, and breast cancer. In addition, scRNA-seq data of immune cells in the TME acquired from human melanoma after immune checkpoint inhibitors were analyzed to investigate the dynamics of glucose metabolic profiles of specific immune cells. RESULTS: Pan-cancer bulk RNA-seq showed that the GLUT3-to-GLUT1 ratio was positively associated with immune cell enrichment score. The scRNA-seq datasets of various human cancer tissues showed that GLUT1 was highly expressed in cancer cells, while GLUT3 was highly expressed in immune cells in TME. The scRNA-seq data obtained from human melanoma tissues pre- and post-immunotherapy showed that glucose metabolism features of myeloid cells, particularly including GLUTs expression, markedly differed according to treatment response. CONCLUSIONS: Differently expressed GLUTs in TME suggest that GLUT could be a good candidate a surrogate of tumor immune metabolic profiles and a target for adjunctive treatments for immunotherapy.
format Online
Article
Text
id pubmed-8710507
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-87105072021-12-28 Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment Choi, Hongyoon Na, Kwon Joong Front Oncol Oncology BACKGROUND: A close metabolic interaction between cancer and immune cells in the tumor microenvironment (TME) plays a pivotal role in cancer immunity. Herein, we have comprehensively investigated the glucose metabolic features of the TME at the single-cell level to discover feasible metabolic targets for the tumor immune status. METHODS: We examined expression levels of glucose transporters (GLUTs) in various cancer types using The Cancer Genome Atlas (TCGA) data and single-cell RNA-seq (scRNA-seq) datasets of human cancer tissues including melanoma, head and neck, and breast cancer. In addition, scRNA-seq data of immune cells in the TME acquired from human melanoma after immune checkpoint inhibitors were analyzed to investigate the dynamics of glucose metabolic profiles of specific immune cells. RESULTS: Pan-cancer bulk RNA-seq showed that the GLUT3-to-GLUT1 ratio was positively associated with immune cell enrichment score. The scRNA-seq datasets of various human cancer tissues showed that GLUT1 was highly expressed in cancer cells, while GLUT3 was highly expressed in immune cells in TME. The scRNA-seq data obtained from human melanoma tissues pre- and post-immunotherapy showed that glucose metabolism features of myeloid cells, particularly including GLUTs expression, markedly differed according to treatment response. CONCLUSIONS: Differently expressed GLUTs in TME suggest that GLUT could be a good candidate a surrogate of tumor immune metabolic profiles and a target for adjunctive treatments for immunotherapy. Frontiers Media S.A. 2021-12-13 /pmc/articles/PMC8710507/ /pubmed/34966676 http://dx.doi.org/10.3389/fonc.2021.769393 Text en Copyright © 2021 Choi and Na https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Choi, Hongyoon
Na, Kwon Joong
Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment
title Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment
title_full Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment
title_fullStr Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment
title_full_unstemmed Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment
title_short Different Glucose Metabolic Features According to Cancer and Immune Cells in the Tumor Microenvironment
title_sort different glucose metabolic features according to cancer and immune cells in the tumor microenvironment
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8710507/
https://www.ncbi.nlm.nih.gov/pubmed/34966676
http://dx.doi.org/10.3389/fonc.2021.769393
work_keys_str_mv AT choihongyoon differentglucosemetabolicfeaturesaccordingtocancerandimmunecellsinthetumormicroenvironment
AT nakwonjoong differentglucosemetabolicfeaturesaccordingtocancerandimmunecellsinthetumormicroenvironment