Cargando…

Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers

BACKGROUND: The main objective of this study was to analyze the effects of KRAS/TP53 mutation status and tumor sideness on the immune microenvironment of colorectal cancer using integrated scRNA-seq data. METHODS: A total of 78 scRNA-seq datasets, comprising 42 treatment-naive colorectal tumors, 13...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Xiaoyu, Xu, Xu, Wu, Zhuozhuo, Shan, Qungang, Wang, Ziyin, Wu, Zhiyuan, Ding, Xiaoyi, Huang, Wei, Wang, Zhongmin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9510840/
https://www.ncbi.nlm.nih.gov/pubmed/36172359
http://dx.doi.org/10.3389/fimmu.2022.961350
_version_ 1784797530401275904
author Liu, Xiaoyu
Xu, Xu
Wu, Zhuozhuo
Shan, Qungang
Wang, Ziyin
Wu, Zhiyuan
Ding, Xiaoyi
Huang, Wei
Wang, Zhongmin
author_facet Liu, Xiaoyu
Xu, Xu
Wu, Zhuozhuo
Shan, Qungang
Wang, Ziyin
Wu, Zhiyuan
Ding, Xiaoyi
Huang, Wei
Wang, Zhongmin
author_sort Liu, Xiaoyu
collection PubMed
description BACKGROUND: The main objective of this study was to analyze the effects of KRAS/TP53 mutation status and tumor sideness on the immune microenvironment of colorectal cancer using integrated scRNA-seq data. METHODS: A total of 78 scRNA-seq datasets, comprising 42 treatment-naive colorectal tumors, 13 tumor adjacent tissues and 23 normal mucosa tissues were included. Standardized Seurat procedures were applied to identify cellular components with canonical cell marks. The batch-effect was assessed and corrected using harmony algorithm. The scMetabolism algorithm was used for single-cell metabolic analysis. The results and clinical significance were further validated using immunofluorescent-staining and TCGA-COAD datasets. Immune-infiltration scores of bulk-RNA-seq data were estimated using ssGSEA. The presto-wilcoxauc algorithm was used to identify differentially enriched genes or pathways across different subgroups. Two-sided p-value less than 0.05 was considered statistically significant. RESULTS: We refined the landscape of functional immune cell subtypes, especially T cells and myeloid cells, across normal mucosa, tumor adjacent and tumor tissue. The existence and function of two states of exhausted CD8(+) T (Tex) subtypes in colorectal cancer, and FOLR2(+) LYVE1(+) macrophages indicating unfavorable prognosis in colorectal cancer were identified and validated. The diverse tumor mutation status reshaped the immune cell function and immune checkpoint ligands/receptors (ICLs/ICRs) expression pattern. Importantly, the KRAS/TP53 dual mutations significantly reduced the major energy metabolic functions in immune cells, and promoted the cell-to-cell communications towards immunosuppression in colorectal cancers. The results revealed LAG3, CD24-SIGLEC10 and HBEGF-CD9 pathways as potential therapeutic targets for dual mutant colorectal cancers. CONCLUSIONS: We revealed that the immune microenvironment underwent a gradual remodeling with an enrichment of immunosuppressive myeloid cells from normal mucosa to tumor regions in colorectal cancers. Moreover, we revealed the metabolic heterogeneity of tumor-infiltrating immune cells and suggested that the KRAS/TP53 dual mutation may impair antitumor immunity by reducing T and myeloid cell energy metabolism and reshaping cellular interactions toward immunosuppression.
format Online
Article
Text
id pubmed-9510840
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-95108402022-09-27 Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers Liu, Xiaoyu Xu, Xu Wu, Zhuozhuo Shan, Qungang Wang, Ziyin Wu, Zhiyuan Ding, Xiaoyi Huang, Wei Wang, Zhongmin Front Immunol Immunology BACKGROUND: The main objective of this study was to analyze the effects of KRAS/TP53 mutation status and tumor sideness on the immune microenvironment of colorectal cancer using integrated scRNA-seq data. METHODS: A total of 78 scRNA-seq datasets, comprising 42 treatment-naive colorectal tumors, 13 tumor adjacent tissues and 23 normal mucosa tissues were included. Standardized Seurat procedures were applied to identify cellular components with canonical cell marks. The batch-effect was assessed and corrected using harmony algorithm. The scMetabolism algorithm was used for single-cell metabolic analysis. The results and clinical significance were further validated using immunofluorescent-staining and TCGA-COAD datasets. Immune-infiltration scores of bulk-RNA-seq data were estimated using ssGSEA. The presto-wilcoxauc algorithm was used to identify differentially enriched genes or pathways across different subgroups. Two-sided p-value less than 0.05 was considered statistically significant. RESULTS: We refined the landscape of functional immune cell subtypes, especially T cells and myeloid cells, across normal mucosa, tumor adjacent and tumor tissue. The existence and function of two states of exhausted CD8(+) T (Tex) subtypes in colorectal cancer, and FOLR2(+) LYVE1(+) macrophages indicating unfavorable prognosis in colorectal cancer were identified and validated. The diverse tumor mutation status reshaped the immune cell function and immune checkpoint ligands/receptors (ICLs/ICRs) expression pattern. Importantly, the KRAS/TP53 dual mutations significantly reduced the major energy metabolic functions in immune cells, and promoted the cell-to-cell communications towards immunosuppression in colorectal cancers. The results revealed LAG3, CD24-SIGLEC10 and HBEGF-CD9 pathways as potential therapeutic targets for dual mutant colorectal cancers. CONCLUSIONS: We revealed that the immune microenvironment underwent a gradual remodeling with an enrichment of immunosuppressive myeloid cells from normal mucosa to tumor regions in colorectal cancers. Moreover, we revealed the metabolic heterogeneity of tumor-infiltrating immune cells and suggested that the KRAS/TP53 dual mutation may impair antitumor immunity by reducing T and myeloid cell energy metabolism and reshaping cellular interactions toward immunosuppression. Frontiers Media S.A. 2022-09-12 /pmc/articles/PMC9510840/ /pubmed/36172359 http://dx.doi.org/10.3389/fimmu.2022.961350 Text en Copyright © 2022 Liu, Xu, Wu, Shan, Wang, Wu, Ding, Huang and Wang 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 Immunology
Liu, Xiaoyu
Xu, Xu
Wu, Zhuozhuo
Shan, Qungang
Wang, Ziyin
Wu, Zhiyuan
Ding, Xiaoyi
Huang, Wei
Wang, Zhongmin
Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers
title Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers
title_full Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers
title_fullStr Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers
title_full_unstemmed Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers
title_short Integrated single-cell RNA-seq analysis identifies immune heterogeneity associated with KRAS/TP53 mutation status and tumor-sideness in colorectal cancers
title_sort integrated single-cell rna-seq analysis identifies immune heterogeneity associated with kras/tp53 mutation status and tumor-sideness in colorectal cancers
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9510840/
https://www.ncbi.nlm.nih.gov/pubmed/36172359
http://dx.doi.org/10.3389/fimmu.2022.961350
work_keys_str_mv AT liuxiaoyu integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT xuxu integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT wuzhuozhuo integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT shanqungang integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT wangziyin integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT wuzhiyuan integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT dingxiaoyi integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT huangwei integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers
AT wangzhongmin integratedsinglecellrnaseqanalysisidentifiesimmuneheterogeneityassociatedwithkrastp53mutationstatusandtumorsidenessincolorectalcancers