Cargando…

Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation

BACKGROUND: The development of atrial fibrillation (AF) following valvular heart disease (VHD) remains a common disease and is associated with substantial adverse complications. However, valid molecular diagnostic and therapeutic tools for post-VHD AF have not been fully established. This study was...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Ao, Jia, Kangni, Liang, Huaibin, Jin, Qi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7948357/
https://www.ncbi.nlm.nih.gov/pubmed/33706714
http://dx.doi.org/10.1186/s12872-021-01939-1
_version_ 1783663391532908544
author Liu, Ao
Jia, Kangni
Liang, Huaibin
Jin, Qi
author_facet Liu, Ao
Jia, Kangni
Liang, Huaibin
Jin, Qi
author_sort Liu, Ao
collection PubMed
description BACKGROUND: The development of atrial fibrillation (AF) following valvular heart disease (VHD) remains a common disease and is associated with substantial adverse complications. However, valid molecular diagnostic and therapeutic tools for post-VHD AF have not been fully established. This study was conducted to discover the molecular mechanisms and immune microenvironment underlying AF following VHD. METHODS: Gene expression profiles of the GSE41177 dataset were assessed to construct a protein–protein interaction network, and then, autophagy-related hub genes were identified. In addition, to determine the functions of immune cell infiltration in valvular AF, we used the CIBERSORT algorithm to estimate the composition of 22 immune cell types in valvular heart disease. Finally, correlation analysis was carried out to identify the relationship between differentially expressed autophagy-related genes (DEARGs) and significant immune cell subpopulations to reveal potential regulatory pathways. RESULTS: A total of 153 DEARGs were identified in AF-VHD patients compared with controlled donors. Moreover, we screened the top ten hub nodes with the highest degrees through a network analysis. The ten hub nodes were considered hub genes related to AF genesis and progression. Then, we revealed six significant immune cell subpopulations through the CIBERSORT algorithm. Finally, correlation analysis was performed, and six DEARGs (BECN1, GAPDH, ATG7, MAPK3, BCL2L1, and MYC) and three immune cell subpopulations (T cells CD4 memory resting, T cells follicular helper, and neutrophils) were identified as the most significant potential regulators. CONCLUSION: The DEARGs and immune cells identified in our study may be critical in AF development following VHD and provide potential predictive markers and therapeutic targets for determining a treatment strategy for AF patients. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12872-021-01939-1.
format Online
Article
Text
id pubmed-7948357
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-79483572021-03-11 Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation Liu, Ao Jia, Kangni Liang, Huaibin Jin, Qi BMC Cardiovasc Disord Research Article BACKGROUND: The development of atrial fibrillation (AF) following valvular heart disease (VHD) remains a common disease and is associated with substantial adverse complications. However, valid molecular diagnostic and therapeutic tools for post-VHD AF have not been fully established. This study was conducted to discover the molecular mechanisms and immune microenvironment underlying AF following VHD. METHODS: Gene expression profiles of the GSE41177 dataset were assessed to construct a protein–protein interaction network, and then, autophagy-related hub genes were identified. In addition, to determine the functions of immune cell infiltration in valvular AF, we used the CIBERSORT algorithm to estimate the composition of 22 immune cell types in valvular heart disease. Finally, correlation analysis was carried out to identify the relationship between differentially expressed autophagy-related genes (DEARGs) and significant immune cell subpopulations to reveal potential regulatory pathways. RESULTS: A total of 153 DEARGs were identified in AF-VHD patients compared with controlled donors. Moreover, we screened the top ten hub nodes with the highest degrees through a network analysis. The ten hub nodes were considered hub genes related to AF genesis and progression. Then, we revealed six significant immune cell subpopulations through the CIBERSORT algorithm. Finally, correlation analysis was performed, and six DEARGs (BECN1, GAPDH, ATG7, MAPK3, BCL2L1, and MYC) and three immune cell subpopulations (T cells CD4 memory resting, T cells follicular helper, and neutrophils) were identified as the most significant potential regulators. CONCLUSION: The DEARGs and immune cells identified in our study may be critical in AF development following VHD and provide potential predictive markers and therapeutic targets for determining a treatment strategy for AF patients. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12872-021-01939-1. BioMed Central 2021-03-11 /pmc/articles/PMC7948357/ /pubmed/33706714 http://dx.doi.org/10.1186/s12872-021-01939-1 Text en © The Author(s) 2021 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/. The Creative Commons Public Domain Dedication waiver (http://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 Article
Liu, Ao
Jia, Kangni
Liang, Huaibin
Jin, Qi
Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation
title Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation
title_full Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation
title_fullStr Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation
title_full_unstemmed Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation
title_short Comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation
title_sort comprehensive analysis of autophagy-related genes and patterns of immune cell infiltration in valvular atrial fibrillation
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7948357/
https://www.ncbi.nlm.nih.gov/pubmed/33706714
http://dx.doi.org/10.1186/s12872-021-01939-1
work_keys_str_mv AT liuao comprehensiveanalysisofautophagyrelatedgenesandpatternsofimmunecellinfiltrationinvalvularatrialfibrillation
AT jiakangni comprehensiveanalysisofautophagyrelatedgenesandpatternsofimmunecellinfiltrationinvalvularatrialfibrillation
AT lianghuaibin comprehensiveanalysisofautophagyrelatedgenesandpatternsofimmunecellinfiltrationinvalvularatrialfibrillation
AT jinqi comprehensiveanalysisofautophagyrelatedgenesandpatternsofimmunecellinfiltrationinvalvularatrialfibrillation