Cargando…

Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis

Mastitis is a common disease in the dairy industry that causes huge economic losses worldwide. Exosomes (carrying proteins, miRNA, lncRNA, etc.) play a vital role in the regulation of immune response. lncRNA can play a variety of regulatory roles by combining with protein, RNA, and DNA. The expressi...

Descripción completa

Detalles Bibliográficos
Autores principales: Chen, Yu, Jing, Hongyuan, Chen, Miaoyu, Liang, Wan, Yang, Jing, Deng, Ganzhen, Guo, Mengyao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8342165/
https://www.ncbi.nlm.nih.gov/pubmed/34367468
http://dx.doi.org/10.1155/2021/8460355
_version_ 1783734010634043392
author Chen, Yu
Jing, Hongyuan
Chen, Miaoyu
Liang, Wan
Yang, Jing
Deng, Ganzhen
Guo, Mengyao
author_facet Chen, Yu
Jing, Hongyuan
Chen, Miaoyu
Liang, Wan
Yang, Jing
Deng, Ganzhen
Guo, Mengyao
author_sort Chen, Yu
collection PubMed
description Mastitis is a common disease in the dairy industry that causes huge economic losses worldwide. Exosomes (carrying proteins, miRNA, lncRNA, etc.) play a vital role in the regulation of immune response. lncRNA can play a variety of regulatory roles by combining with protein, RNA, and DNA. The expression of mRNA and lncRNA in exosomes derived from bovine mammary epithelial cells infected by S. aureus is rarely understood. To explore this issue, RNA sequencing analysis was performed on exosomes derived from S. aureus-infected and noninfected MAC-T cells. Analysis of the sequencing results showed that there were 186 differentially expressed genes, 431 differentially expressed mRNAs and 19 differentially expressed lncRNAs in the exosomes derived from S. aureus-infected and noninfected MAC-T cells. By predicting lncRNA target genes, it was found that 19 differentially expressed lncRNAs all acted on multiple mRNAs in cis and trans. GO analysis revealed that differentially expressed genes and lncRNA target genes played significant roles in such metabolism (reactive oxygen species metabolic processes), transmembrane transport, cellular response to DNA damage stimulus, and response to cytokines. KEGG enrichment indicated that lncRNA target genes gathered in the TNF pathway, Notch pathway, MAPK pathway, NF-kappa B pathway, Hippo pathway, p53 pathway, reactive oxygen species metabolic processes, and longevity regulating pathway. In summary, all data indicated that differentially expressed gene, mRNA, and lncRNA in transcriptional profiling of exosomes participated in bacterial invasion and adhesion, oxidative stress, inflammation, and apoptosis-related signaling pathway. The data obtained in this study would provide valuable resource for understanding the lncRNA information in exosomes derived from dairy cow mammary epithelial cells and conduced to the study of S. aureus infection in dairy cow mammary glands.
format Online
Article
Text
id pubmed-8342165
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-83421652021-08-06 Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis Chen, Yu Jing, Hongyuan Chen, Miaoyu Liang, Wan Yang, Jing Deng, Ganzhen Guo, Mengyao Oxid Med Cell Longev Research Article Mastitis is a common disease in the dairy industry that causes huge economic losses worldwide. Exosomes (carrying proteins, miRNA, lncRNA, etc.) play a vital role in the regulation of immune response. lncRNA can play a variety of regulatory roles by combining with protein, RNA, and DNA. The expression of mRNA and lncRNA in exosomes derived from bovine mammary epithelial cells infected by S. aureus is rarely understood. To explore this issue, RNA sequencing analysis was performed on exosomes derived from S. aureus-infected and noninfected MAC-T cells. Analysis of the sequencing results showed that there were 186 differentially expressed genes, 431 differentially expressed mRNAs and 19 differentially expressed lncRNAs in the exosomes derived from S. aureus-infected and noninfected MAC-T cells. By predicting lncRNA target genes, it was found that 19 differentially expressed lncRNAs all acted on multiple mRNAs in cis and trans. GO analysis revealed that differentially expressed genes and lncRNA target genes played significant roles in such metabolism (reactive oxygen species metabolic processes), transmembrane transport, cellular response to DNA damage stimulus, and response to cytokines. KEGG enrichment indicated that lncRNA target genes gathered in the TNF pathway, Notch pathway, MAPK pathway, NF-kappa B pathway, Hippo pathway, p53 pathway, reactive oxygen species metabolic processes, and longevity regulating pathway. In summary, all data indicated that differentially expressed gene, mRNA, and lncRNA in transcriptional profiling of exosomes participated in bacterial invasion and adhesion, oxidative stress, inflammation, and apoptosis-related signaling pathway. The data obtained in this study would provide valuable resource for understanding the lncRNA information in exosomes derived from dairy cow mammary epithelial cells and conduced to the study of S. aureus infection in dairy cow mammary glands. Hindawi 2021-07-29 /pmc/articles/PMC8342165/ /pubmed/34367468 http://dx.doi.org/10.1155/2021/8460355 Text en Copyright © 2021 Yu Chen et al. https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Chen, Yu
Jing, Hongyuan
Chen, Miaoyu
Liang, Wan
Yang, Jing
Deng, Ganzhen
Guo, Mengyao
Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis
title Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis
title_full Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis
title_fullStr Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis
title_full_unstemmed Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis
title_short Transcriptional Profiling of Exosomes Derived from Staphylococcus aureus-Infected Bovine Mammary Epithelial Cell Line MAC-T by RNA-Seq Analysis
title_sort transcriptional profiling of exosomes derived from staphylococcus aureus-infected bovine mammary epithelial cell line mac-t by rna-seq analysis
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8342165/
https://www.ncbi.nlm.nih.gov/pubmed/34367468
http://dx.doi.org/10.1155/2021/8460355
work_keys_str_mv AT chenyu transcriptionalprofilingofexosomesderivedfromstaphylococcusaureusinfectedbovinemammaryepithelialcelllinemactbyrnaseqanalysis
AT jinghongyuan transcriptionalprofilingofexosomesderivedfromstaphylococcusaureusinfectedbovinemammaryepithelialcelllinemactbyrnaseqanalysis
AT chenmiaoyu transcriptionalprofilingofexosomesderivedfromstaphylococcusaureusinfectedbovinemammaryepithelialcelllinemactbyrnaseqanalysis
AT liangwan transcriptionalprofilingofexosomesderivedfromstaphylococcusaureusinfectedbovinemammaryepithelialcelllinemactbyrnaseqanalysis
AT yangjing transcriptionalprofilingofexosomesderivedfromstaphylococcusaureusinfectedbovinemammaryepithelialcelllinemactbyrnaseqanalysis
AT dengganzhen transcriptionalprofilingofexosomesderivedfromstaphylococcusaureusinfectedbovinemammaryepithelialcelllinemactbyrnaseqanalysis
AT guomengyao transcriptionalprofilingofexosomesderivedfromstaphylococcusaureusinfectedbovinemammaryepithelialcelllinemactbyrnaseqanalysis