Cargando…

Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis

Haemophilus parasuis is the causative agent of Glässer’s disease in pigs. H. parasuis can cause vascular damage, although the mechanism remains unclear. In this study, we investigated the host cell responses involved in the molecular pathway interactions in porcine aortic vascular endothelial cells...

Descripción completa

Detalles Bibliográficos
Autores principales: Fu, Shulin, Guo, Jing, Li, Ruizhi, Qiu, Yinsheng, Ye, Chun, Liu, Yu, Wu, Zhongyuan, Guo, Ling, Hou, Yongqing, Hu, Chien-An Andy
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5983834/
https://www.ncbi.nlm.nih.gov/pubmed/29710817
http://dx.doi.org/10.3390/ijms19051320
_version_ 1783328510514823168
author Fu, Shulin
Guo, Jing
Li, Ruizhi
Qiu, Yinsheng
Ye, Chun
Liu, Yu
Wu, Zhongyuan
Guo, Ling
Hou, Yongqing
Hu, Chien-An Andy
author_facet Fu, Shulin
Guo, Jing
Li, Ruizhi
Qiu, Yinsheng
Ye, Chun
Liu, Yu
Wu, Zhongyuan
Guo, Ling
Hou, Yongqing
Hu, Chien-An Andy
author_sort Fu, Shulin
collection PubMed
description Haemophilus parasuis is the causative agent of Glässer’s disease in pigs. H. parasuis can cause vascular damage, although the mechanism remains unclear. In this study, we investigated the host cell responses involved in the molecular pathway interactions in porcine aortic vascular endothelial cells (PAVECs) induced by H. parasuis using RNA-Seq. The transcriptome results showed that when PAVECs were infected with H. parasuis for 24 h, 281 differentially expressed genes (DEGs) were identified; of which, 236 were upregulated and 45 downregulated. The 281 DEGs were involved in 136 KEGG signaling pathways that were organismal systems, environmental information processing, metabolism, cellular processes, and genetic information processing. The main pathways were the Rap1, FoxO, and PI3K/Akt signaling pathways, and the overexpressed genes were determined and verified by quantitative reverse transcription polymerase chain reaction. In addition, 252 genes were clustered into biological processes, molecular processes, and cellular components. Our study provides new insights for understanding the interaction between bacterial and host cells, and analyzed, in detail, the possible mechanisms that lead to vascular damage induced by H. parasuis. This may lead to development of novel therapeutic targets to control H. parasuis infection.
format Online
Article
Text
id pubmed-5983834
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-59838342018-06-05 Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis Fu, Shulin Guo, Jing Li, Ruizhi Qiu, Yinsheng Ye, Chun Liu, Yu Wu, Zhongyuan Guo, Ling Hou, Yongqing Hu, Chien-An Andy Int J Mol Sci Article Haemophilus parasuis is the causative agent of Glässer’s disease in pigs. H. parasuis can cause vascular damage, although the mechanism remains unclear. In this study, we investigated the host cell responses involved in the molecular pathway interactions in porcine aortic vascular endothelial cells (PAVECs) induced by H. parasuis using RNA-Seq. The transcriptome results showed that when PAVECs were infected with H. parasuis for 24 h, 281 differentially expressed genes (DEGs) were identified; of which, 236 were upregulated and 45 downregulated. The 281 DEGs were involved in 136 KEGG signaling pathways that were organismal systems, environmental information processing, metabolism, cellular processes, and genetic information processing. The main pathways were the Rap1, FoxO, and PI3K/Akt signaling pathways, and the overexpressed genes were determined and verified by quantitative reverse transcription polymerase chain reaction. In addition, 252 genes were clustered into biological processes, molecular processes, and cellular components. Our study provides new insights for understanding the interaction between bacterial and host cells, and analyzed, in detail, the possible mechanisms that lead to vascular damage induced by H. parasuis. This may lead to development of novel therapeutic targets to control H. parasuis infection. MDPI 2018-04-29 /pmc/articles/PMC5983834/ /pubmed/29710817 http://dx.doi.org/10.3390/ijms19051320 Text en © 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Fu, Shulin
Guo, Jing
Li, Ruizhi
Qiu, Yinsheng
Ye, Chun
Liu, Yu
Wu, Zhongyuan
Guo, Ling
Hou, Yongqing
Hu, Chien-An Andy
Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis
title Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis
title_full Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis
title_fullStr Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis
title_full_unstemmed Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis
title_short Transcriptional Profiling of Host Cell Responses to Virulent Haemophilus parasuis: New Insights into Pathogenesis
title_sort transcriptional profiling of host cell responses to virulent haemophilus parasuis: new insights into pathogenesis
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5983834/
https://www.ncbi.nlm.nih.gov/pubmed/29710817
http://dx.doi.org/10.3390/ijms19051320
work_keys_str_mv AT fushulin transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT guojing transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT liruizhi transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT qiuyinsheng transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT yechun transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT liuyu transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT wuzhongyuan transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT guoling transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT houyongqing transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis
AT huchienanandy transcriptionalprofilingofhostcellresponsestovirulenthaemophilusparasuisnewinsightsintopathogenesis