Cargando…

A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma

BACKGROUND AND AIM: Lipid metabolic reprogramming is considered to be a new hallmark of malignant tumors. The purpose of this study was to explore the expression profiles of lipid metabolism-related genes (LMRG) in colorectal cancer (CRC). METHODS: The lipid metabolism statuses of 500 CRC patients f...

Descripción completa

Detalles Bibliográficos
Autores principales: Yang, Chao, Huang, Shuoyang, Cao, Fengyu, Zheng, Yongbin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8571885/
https://www.ncbi.nlm.nih.gov/pubmed/34740325
http://dx.doi.org/10.1186/s12885-021-08902-5
_version_ 1784595110140313600
author Yang, Chao
Huang, Shuoyang
Cao, Fengyu
Zheng, Yongbin
author_facet Yang, Chao
Huang, Shuoyang
Cao, Fengyu
Zheng, Yongbin
author_sort Yang, Chao
collection PubMed
description BACKGROUND AND AIM: Lipid metabolic reprogramming is considered to be a new hallmark of malignant tumors. The purpose of this study was to explore the expression profiles of lipid metabolism-related genes (LMRG) in colorectal cancer (CRC). METHODS: The lipid metabolism statuses of 500 CRC patients from the Cancer Genome Atlas (TCGA) and 523 from the Gene Expression Omnibus (GEO GSE39582) database were analyzed. The risk signature was constructed by univariate Cox regression and least absolute shrinkage and selection operator (LASSO) Cox regression. RESULTS: A novel four-LMRG signature (PROCA1, CCKBR, CPT2, and FDFT1) was constructed to predict clinical outcomes in CRC patients. The risk signature was shown to be an independent prognostic factor for CRC and was associated with tumour malignancy. Principal components analysis demonstrated that the risk signature could distinguish between low- and high-risk patients. There were significantly differences in abundances of tumor-infiltrating immune cells and mutational landscape between the two risk groups. Patients in the low-risk group were more likely to have higher tumor mutational burden, stem cell characteristics, and higher PD-L1 expression levels. Furthermore, a genomic-clinicopathologic nomogram was established and shown to be a more effective risk stratification tool than any clinical parameter alone. CONCLUSIONS: This study demonstrated the prognostic value of LMRG and showed that they may be partially involved in the suppressive immune microenvironment formation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12885-021-08902-5.
format Online
Article
Text
id pubmed-8571885
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-85718852021-11-08 A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma Yang, Chao Huang, Shuoyang Cao, Fengyu Zheng, Yongbin BMC Cancer Research BACKGROUND AND AIM: Lipid metabolic reprogramming is considered to be a new hallmark of malignant tumors. The purpose of this study was to explore the expression profiles of lipid metabolism-related genes (LMRG) in colorectal cancer (CRC). METHODS: The lipid metabolism statuses of 500 CRC patients from the Cancer Genome Atlas (TCGA) and 523 from the Gene Expression Omnibus (GEO GSE39582) database were analyzed. The risk signature was constructed by univariate Cox regression and least absolute shrinkage and selection operator (LASSO) Cox regression. RESULTS: A novel four-LMRG signature (PROCA1, CCKBR, CPT2, and FDFT1) was constructed to predict clinical outcomes in CRC patients. The risk signature was shown to be an independent prognostic factor for CRC and was associated with tumour malignancy. Principal components analysis demonstrated that the risk signature could distinguish between low- and high-risk patients. There were significantly differences in abundances of tumor-infiltrating immune cells and mutational landscape between the two risk groups. Patients in the low-risk group were more likely to have higher tumor mutational burden, stem cell characteristics, and higher PD-L1 expression levels. Furthermore, a genomic-clinicopathologic nomogram was established and shown to be a more effective risk stratification tool than any clinical parameter alone. CONCLUSIONS: This study demonstrated the prognostic value of LMRG and showed that they may be partially involved in the suppressive immune microenvironment formation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12885-021-08902-5. BioMed Central 2021-11-05 /pmc/articles/PMC8571885/ /pubmed/34740325 http://dx.doi.org/10.1186/s12885-021-08902-5 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Yang, Chao
Huang, Shuoyang
Cao, Fengyu
Zheng, Yongbin
A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma
title A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma
title_full A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma
title_fullStr A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma
title_full_unstemmed A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma
title_short A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma
title_sort lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8571885/
https://www.ncbi.nlm.nih.gov/pubmed/34740325
http://dx.doi.org/10.1186/s12885-021-08902-5
work_keys_str_mv AT yangchao alipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma
AT huangshuoyang alipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma
AT caofengyu alipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma
AT zhengyongbin alipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma
AT yangchao lipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma
AT huangshuoyang lipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma
AT caofengyu lipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma
AT zhengyongbin lipidmetabolismrelatedgenesprognosisbiomarkerassociatedwiththetumorimmunemicroenvironmentincolorectalcarcinoma