Cargando…

Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish

The Toll/interleukin-1 receptor (TIR) domain is a structural unit responsible for the assembly of signal protein complexes in Toll-like receptor (TLR) and interleukin-1 receptor signaling pathways. TIR domain homologs are found in a considerable number of bacteria and enhance bacterial infection and...

Descripción completa

Detalles Bibliográficos
Autores principales: Tang, Huai-ping, Huang, Chen, Hu, Chong-bin, Li, Hao, Shao, Tong, Ji, Jian-fei, Bai, Jun, Fan, Dong-dong, Lin, Ai-fu, Xiang, Li-xin, Shao, Jian-zhong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8297594/
https://www.ncbi.nlm.nih.gov/pubmed/34305858
http://dx.doi.org/10.3389/fmicb.2021.694081
_version_ 1783725888499613696
author Tang, Huai-ping
Huang, Chen
Hu, Chong-bin
Li, Hao
Shao, Tong
Ji, Jian-fei
Bai, Jun
Fan, Dong-dong
Lin, Ai-fu
Xiang, Li-xin
Shao, Jian-zhong
author_facet Tang, Huai-ping
Huang, Chen
Hu, Chong-bin
Li, Hao
Shao, Tong
Ji, Jian-fei
Bai, Jun
Fan, Dong-dong
Lin, Ai-fu
Xiang, Li-xin
Shao, Jian-zhong
author_sort Tang, Huai-ping
collection PubMed
description The Toll/interleukin-1 receptor (TIR) domain is a structural unit responsible for the assembly of signal protein complexes in Toll-like receptor (TLR) and interleukin-1 receptor signaling pathways. TIR domain homologs are found in a considerable number of bacteria and enhance bacterial infection and survival in host organisms. However, whether TIR domain homologs exist in Aeromonas hydrophila, a ubiquitous waterborne bacterium in aquatic environments, remains poorly understood. In this study, a TIR domain protein (TcpAh) was identified from A. hydrophila JBN2301. TIR domain of TcpAh is highly homologous to the counterpart domains in TLRs and myeloid differentiation factor 88 (MyD88). The zebrafish infected with mutant A. hydrophila with tcpAh deletion had a remarkably lower mortality than those infected with the wild-type strain. This result suggests that TcpAh is a crucial virulence factor for A. hydrophila infection. TcpAh exhibited a strong ability to associate with MyD88, tumor necrosis factor receptor-associated factor 3 (TRAF3) and TRAF-associated NF-κB activator-binding kinase 1 (TBK1) in TIR–TIR, TIR–Death domain (DD), and other alternative interactions. This finding suggests that TcpAh extensively interferes with MyD88 and TIR domain-containing adapter inducing interferon (IFN)-β (TRIF) signaling pathways downstream of TLRs. Consequently, CD80/86 expression was suppressed by TcpAh via attenuating TLR-stimulated NF-κB activation, which ultimately led to the impairment of the major costimulatory signal essential for the initiation of adaptive humoral immunity against A. hydrophila infection. We believe that this study is the first to show a previously unrecognized mechanism underlying A. hydrophila evades from host antibacterial defense by intervening CD80/86 signal, which bridges innate and adaptive immunity. The mechanism will benefit the development of therapeutic interventions for A. hydrophila infection and septicemia by targeting TcpAh homologs.
format Online
Article
Text
id pubmed-8297594
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-82975942021-07-23 Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish Tang, Huai-ping Huang, Chen Hu, Chong-bin Li, Hao Shao, Tong Ji, Jian-fei Bai, Jun Fan, Dong-dong Lin, Ai-fu Xiang, Li-xin Shao, Jian-zhong Front Microbiol Microbiology The Toll/interleukin-1 receptor (TIR) domain is a structural unit responsible for the assembly of signal protein complexes in Toll-like receptor (TLR) and interleukin-1 receptor signaling pathways. TIR domain homologs are found in a considerable number of bacteria and enhance bacterial infection and survival in host organisms. However, whether TIR domain homologs exist in Aeromonas hydrophila, a ubiquitous waterborne bacterium in aquatic environments, remains poorly understood. In this study, a TIR domain protein (TcpAh) was identified from A. hydrophila JBN2301. TIR domain of TcpAh is highly homologous to the counterpart domains in TLRs and myeloid differentiation factor 88 (MyD88). The zebrafish infected with mutant A. hydrophila with tcpAh deletion had a remarkably lower mortality than those infected with the wild-type strain. This result suggests that TcpAh is a crucial virulence factor for A. hydrophila infection. TcpAh exhibited a strong ability to associate with MyD88, tumor necrosis factor receptor-associated factor 3 (TRAF3) and TRAF-associated NF-κB activator-binding kinase 1 (TBK1) in TIR–TIR, TIR–Death domain (DD), and other alternative interactions. This finding suggests that TcpAh extensively interferes with MyD88 and TIR domain-containing adapter inducing interferon (IFN)-β (TRIF) signaling pathways downstream of TLRs. Consequently, CD80/86 expression was suppressed by TcpAh via attenuating TLR-stimulated NF-κB activation, which ultimately led to the impairment of the major costimulatory signal essential for the initiation of adaptive humoral immunity against A. hydrophila infection. We believe that this study is the first to show a previously unrecognized mechanism underlying A. hydrophila evades from host antibacterial defense by intervening CD80/86 signal, which bridges innate and adaptive immunity. The mechanism will benefit the development of therapeutic interventions for A. hydrophila infection and septicemia by targeting TcpAh homologs. Frontiers Media S.A. 2021-07-08 /pmc/articles/PMC8297594/ /pubmed/34305858 http://dx.doi.org/10.3389/fmicb.2021.694081 Text en Copyright © 2021 Tang, Huang, Hu, Li, Shao, Ji, Bai, Fan, Lin, Xiang and Shao. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Microbiology
Tang, Huai-ping
Huang, Chen
Hu, Chong-bin
Li, Hao
Shao, Tong
Ji, Jian-fei
Bai, Jun
Fan, Dong-dong
Lin, Ai-fu
Xiang, Li-xin
Shao, Jian-zhong
Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish
title Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish
title_full Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish
title_fullStr Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish
title_full_unstemmed Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish
title_short Inhibitory Role of an Aeromonas hydrophila TIR Domain Effector in Antibacterial Immunity by Targeting TLR Signaling Complexes in Zebrafish
title_sort inhibitory role of an aeromonas hydrophila tir domain effector in antibacterial immunity by targeting tlr signaling complexes in zebrafish
topic Microbiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8297594/
https://www.ncbi.nlm.nih.gov/pubmed/34305858
http://dx.doi.org/10.3389/fmicb.2021.694081
work_keys_str_mv AT tanghuaiping inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT huangchen inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT huchongbin inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT lihao inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT shaotong inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT jijianfei inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT baijun inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT fandongdong inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT linaifu inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT xianglixin inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish
AT shaojianzhong inhibitoryroleofanaeromonashydrophilatirdomaineffectorinantibacterialimmunitybytargetingtlrsignalingcomplexesinzebrafish