Cargando…

An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice

Rabies remains an important public health threat, killing approximately 59,000 people worldwide annually, most of which are from the developing countries of Africa and Asia where dog rabies are endemic. Therefore, developing an affordable and efficacious vaccine for dog-mediated rabies control is ne...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Zhao, Liang, Qian, Zhang, Yajing, Yang, Jie, Li, Mingming, Wang, Kunlun, Cui, Min, Chen, Huanchun, Fu, Zhen F., Zhao, Ling, Zhou, Ming
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5663534/
https://www.ncbi.nlm.nih.gov/pubmed/29137362
http://dx.doi.org/10.18632/oncotarget.18368
_version_ 1783274826764386304
author Wang, Zhao
Liang, Qian
Zhang, Yajing
Yang, Jie
Li, Mingming
Wang, Kunlun
Cui, Min
Chen, Huanchun
Fu, Zhen F.
Zhao, Ling
Zhou, Ming
author_facet Wang, Zhao
Liang, Qian
Zhang, Yajing
Yang, Jie
Li, Mingming
Wang, Kunlun
Cui, Min
Chen, Huanchun
Fu, Zhen F.
Zhao, Ling
Zhou, Ming
author_sort Wang, Zhao
collection PubMed
description Rabies remains an important public health threat, killing approximately 59,000 people worldwide annually, most of which are from the developing countries of Africa and Asia where dog rabies are endemic. Therefore, developing an affordable and efficacious vaccine for dog-mediated rabies control is needful in these countries. Our previous studies indicated that over-expression of granulocyte-macrophage colony stimulating factor (GM-CSF) or macrophage inflammatory protein-1 (MIP-1α or CCL3) by recombinant rabies virus (rRABV) could enhance the immunogenicity by activating dendritic cells (DCs). In this study, to further characterize the role of activating DCs in RABV immunogenicity, High mobility group box 1 (HMGB1), a highly conserved and non-histone chromosomal protein that can promote DCs maturation and activation, were investigated. The wild-type HMGB1 (HMGB1(wt)) and an optimized HMGB1 (HMGB1(mut)) were individually inserted into the genome of the rRABV strain LBNSE (designated as LBNSE-HMGB1(wt) and LBNSE-HMGB1(mut), respectively), and the effect of over-expression of HMGB1 on the immunogenicity of RABV was investigated. The results demonstrated that LBNSE-HMGB1(mut) could promote significantly more DCs activation, and the recruitment of follicular helper T, germinal center B and plasma cells in vaccinated mice than those immunized with LBNSE-HMGB1(wt) or parent virus LBNSE. Further investigations suggested that mice vaccinated with LBNSE-HMGB1(mut) produced significantly higher level of RABV-neutralizing antibodies and offered a better protection than those vaccinated with LBNSE or LBNSE-HMGB1(wt). Taken together, these data provides a better understanding of the mechanism for HMGB1 as a potential adjuvant in enhancing the immunogenicity of RABV, which would contribute to developing more-efficacious rabies vaccines.
format Online
Article
Text
id pubmed-5663534
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-56635342017-11-13 An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice Wang, Zhao Liang, Qian Zhang, Yajing Yang, Jie Li, Mingming Wang, Kunlun Cui, Min Chen, Huanchun Fu, Zhen F. Zhao, Ling Zhou, Ming Oncotarget Research Paper Rabies remains an important public health threat, killing approximately 59,000 people worldwide annually, most of which are from the developing countries of Africa and Asia where dog rabies are endemic. Therefore, developing an affordable and efficacious vaccine for dog-mediated rabies control is needful in these countries. Our previous studies indicated that over-expression of granulocyte-macrophage colony stimulating factor (GM-CSF) or macrophage inflammatory protein-1 (MIP-1α or CCL3) by recombinant rabies virus (rRABV) could enhance the immunogenicity by activating dendritic cells (DCs). In this study, to further characterize the role of activating DCs in RABV immunogenicity, High mobility group box 1 (HMGB1), a highly conserved and non-histone chromosomal protein that can promote DCs maturation and activation, were investigated. The wild-type HMGB1 (HMGB1(wt)) and an optimized HMGB1 (HMGB1(mut)) were individually inserted into the genome of the rRABV strain LBNSE (designated as LBNSE-HMGB1(wt) and LBNSE-HMGB1(mut), respectively), and the effect of over-expression of HMGB1 on the immunogenicity of RABV was investigated. The results demonstrated that LBNSE-HMGB1(mut) could promote significantly more DCs activation, and the recruitment of follicular helper T, germinal center B and plasma cells in vaccinated mice than those immunized with LBNSE-HMGB1(wt) or parent virus LBNSE. Further investigations suggested that mice vaccinated with LBNSE-HMGB1(mut) produced significantly higher level of RABV-neutralizing antibodies and offered a better protection than those vaccinated with LBNSE or LBNSE-HMGB1(wt). Taken together, these data provides a better understanding of the mechanism for HMGB1 as a potential adjuvant in enhancing the immunogenicity of RABV, which would contribute to developing more-efficacious rabies vaccines. Impact Journals LLC 2017-06-05 /pmc/articles/PMC5663534/ /pubmed/29137362 http://dx.doi.org/10.18632/oncotarget.18368 Text en Copyright: © 2017 Wang et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (http://creativecommons.org/licenses/by/3.0/) (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Wang, Zhao
Liang, Qian
Zhang, Yajing
Yang, Jie
Li, Mingming
Wang, Kunlun
Cui, Min
Chen, Huanchun
Fu, Zhen F.
Zhao, Ling
Zhou, Ming
An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice
title An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice
title_full An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice
title_fullStr An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice
title_full_unstemmed An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice
title_short An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice
title_sort optimized hmgb1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5663534/
https://www.ncbi.nlm.nih.gov/pubmed/29137362
http://dx.doi.org/10.18632/oncotarget.18368
work_keys_str_mv AT wangzhao anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT liangqian anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT zhangyajing anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT yangjie anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT limingming anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT wangkunlun anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT cuimin anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT chenhuanchun anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT fuzhenf anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT zhaoling anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT zhouming anoptimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT wangzhao optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT liangqian optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT zhangyajing optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT yangjie optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT limingming optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT wangkunlun optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT cuimin optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT chenhuanchun optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT fuzhenf optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT zhaoling optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice
AT zhouming optimizedhmgb1expressedbyrecombinantrabiesvirusenhancesimmunogenicitythroughactivationofdendriticcellsinmice