Cargando…

Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm

AIM: Autophagy plays essential roles in abdominal aortic aneurysm (AAA) development and progression. The objective of this study was to verify the autophagy-related genes (ARGs) underlying AAA empirically and using bioinformatics analysis. METHODS: Two gene expression profile datasets GSE98278 and G...

Descripción completa

Detalles Bibliográficos
Autores principales: Yuan, Xiaoli, Song, Yancheng, Xin, Hai, Zhang, Lu, Liu, Bingyu, Ma, Jianmin, Sun, Ruicong, Guan, Xiaomei, Jiang, Zhirong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10515431/
https://www.ncbi.nlm.nih.gov/pubmed/37737183
http://dx.doi.org/10.1186/s40001-023-01354-6
_version_ 1785108946964447232
author Yuan, Xiaoli
Song, Yancheng
Xin, Hai
Zhang, Lu
Liu, Bingyu
Ma, Jianmin
Sun, Ruicong
Guan, Xiaomei
Jiang, Zhirong
author_facet Yuan, Xiaoli
Song, Yancheng
Xin, Hai
Zhang, Lu
Liu, Bingyu
Ma, Jianmin
Sun, Ruicong
Guan, Xiaomei
Jiang, Zhirong
author_sort Yuan, Xiaoli
collection PubMed
description AIM: Autophagy plays essential roles in abdominal aortic aneurysm (AAA) development and progression. The objective of this study was to verify the autophagy-related genes (ARGs) underlying AAA empirically and using bioinformatics analysis. METHODS: Two gene expression profile datasets GSE98278 and GSE57691 were downloaded from the Gene Expression Omnibus (GEO) database, and principal component analysis was performed. Following, the R software (version 4.0.0) was employed to analyze potentially differentially expressed genes related with AAA and autophagy. Subsequently, the candidate genes were screened using protein–protein interaction (PPI), gene ontology (GO) enrichment analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Finally, quantitative real-time polymerase chain reaction (RT-qPCR) was performed to detect the RNA expression levels of the top five selected abnormal ARGs in clinical samples obtained from the normal and AAA patients. RESULTS: According to the information contained (97 AAA patients and 10 healthy controls) in the two datasets, a total of 44 differentially expressed autophagy-related genes (6 up-regulated genes and 38 down-regulated genes) were screened. GO enrichment analysis of differentially expressed autophagy-related genes (DEARGs) demonstrated that some enrichment items were associated with inflammation, and PPI analysis indicated interaction between these genes. RT-qPCR results presented that the expression levels of IL6, PPARG, SOD1, and MAP1LC3B were in accordance with the bioinformatics prediction results acquired from the mRNA chip. CONCLUSION: Bioinformatics analysis identified 44 potential autophagy-related differentially expressed genes in AAA. Further verification by RT- qPCR presented that IL6, PPARG, SOD1, and MAP1LC3B may affect the development of AAA by regulating autophagy. These findings might help explain the pathogenesis of AAA and be helpful in its diagnosis and treatment.
format Online
Article
Text
id pubmed-10515431
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-105154312023-09-23 Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm Yuan, Xiaoli Song, Yancheng Xin, Hai Zhang, Lu Liu, Bingyu Ma, Jianmin Sun, Ruicong Guan, Xiaomei Jiang, Zhirong Eur J Med Res Research AIM: Autophagy plays essential roles in abdominal aortic aneurysm (AAA) development and progression. The objective of this study was to verify the autophagy-related genes (ARGs) underlying AAA empirically and using bioinformatics analysis. METHODS: Two gene expression profile datasets GSE98278 and GSE57691 were downloaded from the Gene Expression Omnibus (GEO) database, and principal component analysis was performed. Following, the R software (version 4.0.0) was employed to analyze potentially differentially expressed genes related with AAA and autophagy. Subsequently, the candidate genes were screened using protein–protein interaction (PPI), gene ontology (GO) enrichment analysis, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Finally, quantitative real-time polymerase chain reaction (RT-qPCR) was performed to detect the RNA expression levels of the top five selected abnormal ARGs in clinical samples obtained from the normal and AAA patients. RESULTS: According to the information contained (97 AAA patients and 10 healthy controls) in the two datasets, a total of 44 differentially expressed autophagy-related genes (6 up-regulated genes and 38 down-regulated genes) were screened. GO enrichment analysis of differentially expressed autophagy-related genes (DEARGs) demonstrated that some enrichment items were associated with inflammation, and PPI analysis indicated interaction between these genes. RT-qPCR results presented that the expression levels of IL6, PPARG, SOD1, and MAP1LC3B were in accordance with the bioinformatics prediction results acquired from the mRNA chip. CONCLUSION: Bioinformatics analysis identified 44 potential autophagy-related differentially expressed genes in AAA. Further verification by RT- qPCR presented that IL6, PPARG, SOD1, and MAP1LC3B may affect the development of AAA by regulating autophagy. These findings might help explain the pathogenesis of AAA and be helpful in its diagnosis and treatment. BioMed Central 2023-09-22 /pmc/articles/PMC10515431/ /pubmed/37737183 http://dx.doi.org/10.1186/s40001-023-01354-6 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This 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
Yuan, Xiaoli
Song, Yancheng
Xin, Hai
Zhang, Lu
Liu, Bingyu
Ma, Jianmin
Sun, Ruicong
Guan, Xiaomei
Jiang, Zhirong
Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm
title Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm
title_full Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm
title_fullStr Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm
title_full_unstemmed Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm
title_short Identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm
title_sort identification and experimental validation of autophagy-related genes in abdominal aortic aneurysm
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10515431/
https://www.ncbi.nlm.nih.gov/pubmed/37737183
http://dx.doi.org/10.1186/s40001-023-01354-6
work_keys_str_mv AT yuanxiaoli identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT songyancheng identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT xinhai identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT zhanglu identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT liubingyu identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT majianmin identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT sunruicong identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT guanxiaomei identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm
AT jiangzhirong identificationandexperimentalvalidationofautophagyrelatedgenesinabdominalaorticaneurysm