Cargando…

Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease

Rabies virus is a highly neurovirulent RNA virus, which causes about 59000 deaths in humans each year. Previously, we described macrophage cytotoxicity upon infection with rabies virus. Here we examined the type of cell death and the role of specific caspases in cell death and disease development up...

Descripción completa

Detalles Bibliográficos
Autores principales: Kip, E, Nazé, F, Suin, V, Vanden Berghe, T, Francart, A, Lamoral, S, Vandenabeele, P, Beyaert, R, Van Gucht, S, Kalai, M
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5339016/
https://www.ncbi.nlm.nih.gov/pubmed/28280602
http://dx.doi.org/10.1038/cddiscovery.2017.12
_version_ 1782512605102866432
author Kip, E
Nazé, F
Suin, V
Vanden Berghe, T
Francart, A
Lamoral, S
Vandenabeele, P
Beyaert, R
Van Gucht, S
Kalai, M
author_facet Kip, E
Nazé, F
Suin, V
Vanden Berghe, T
Francart, A
Lamoral, S
Vandenabeele, P
Beyaert, R
Van Gucht, S
Kalai, M
author_sort Kip, E
collection PubMed
description Rabies virus is a highly neurovirulent RNA virus, which causes about 59000 deaths in humans each year. Previously, we described macrophage cytotoxicity upon infection with rabies virus. Here we examined the type of cell death and the role of specific caspases in cell death and disease development upon infection with two laboratory strains of rabies virus: Challenge Virus Standard strain-11 (CVS-11) is highly neurotropic and lethal for mice, while the attenuated Evelyn–Rotnycki–Abelseth (ERA) strain has a broader cell tropism, is non-lethal and has been used as an oral vaccine for animals. Infection of Mf4/4 macrophages with both strains led to caspase-1 activation and IL-1β and IL-18 production, as well as activation of caspases-3, -7, -8, and -9. Moreover, absence of caspase-3, but not of caspase-1 and -11 or -7, partially inhibited virus-induced cell death of bone marrow-derived macrophages. Intranasal inoculation with CVS-11 of mice deficient for either caspase-1 and -11 or -7 or both IL-1β and IL-18 led to general brain infection and lethal disease similar to wild-type mice. Deficiency of caspase-3, on the other hand, significantly delayed the onset of disease, but did not prevent final lethal outcome. Interestingly, deficiency of caspase-1/11, the key executioner of pyroptosis, aggravated disease severity caused by ERA virus, whereas wild-type mice or mice deficient for either caspase-3, -7, or both IL-1β and IL-18 presented the typical mild symptoms associated with ERA virus. In conclusion, rabies virus infection of macrophages induces caspase-1- and caspase-3-dependent cell death. In vivo caspase-1/11 and caspase-3 differently affect disease development in response to infection with the attenuated ERA strain or the virulent CVS-11 strain, respectively. Inflammatory caspases seem to control attenuated rabies virus infection, while caspase-3 aggravates virulent rabies virus infection.
format Online
Article
Text
id pubmed-5339016
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-53390162017-03-09 Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease Kip, E Nazé, F Suin, V Vanden Berghe, T Francart, A Lamoral, S Vandenabeele, P Beyaert, R Van Gucht, S Kalai, M Cell Death Discov Article Rabies virus is a highly neurovirulent RNA virus, which causes about 59000 deaths in humans each year. Previously, we described macrophage cytotoxicity upon infection with rabies virus. Here we examined the type of cell death and the role of specific caspases in cell death and disease development upon infection with two laboratory strains of rabies virus: Challenge Virus Standard strain-11 (CVS-11) is highly neurotropic and lethal for mice, while the attenuated Evelyn–Rotnycki–Abelseth (ERA) strain has a broader cell tropism, is non-lethal and has been used as an oral vaccine for animals. Infection of Mf4/4 macrophages with both strains led to caspase-1 activation and IL-1β and IL-18 production, as well as activation of caspases-3, -7, -8, and -9. Moreover, absence of caspase-3, but not of caspase-1 and -11 or -7, partially inhibited virus-induced cell death of bone marrow-derived macrophages. Intranasal inoculation with CVS-11 of mice deficient for either caspase-1 and -11 or -7 or both IL-1β and IL-18 led to general brain infection and lethal disease similar to wild-type mice. Deficiency of caspase-3, on the other hand, significantly delayed the onset of disease, but did not prevent final lethal outcome. Interestingly, deficiency of caspase-1/11, the key executioner of pyroptosis, aggravated disease severity caused by ERA virus, whereas wild-type mice or mice deficient for either caspase-3, -7, or both IL-1β and IL-18 presented the typical mild symptoms associated with ERA virus. In conclusion, rabies virus infection of macrophages induces caspase-1- and caspase-3-dependent cell death. In vivo caspase-1/11 and caspase-3 differently affect disease development in response to infection with the attenuated ERA strain or the virulent CVS-11 strain, respectively. Inflammatory caspases seem to control attenuated rabies virus infection, while caspase-3 aggravates virulent rabies virus infection. Nature Publishing Group 2017-03-06 /pmc/articles/PMC5339016/ /pubmed/28280602 http://dx.doi.org/10.1038/cddiscovery.2017.12 Text en Copyright © 2017 The Author(s) http://creativecommons.org/licenses/by/4.0/ This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Article
Kip, E
Nazé, F
Suin, V
Vanden Berghe, T
Francart, A
Lamoral, S
Vandenabeele, P
Beyaert, R
Van Gucht, S
Kalai, M
Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease
title Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease
title_full Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease
title_fullStr Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease
title_full_unstemmed Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease
title_short Impact of caspase-1/11, -3, -7, or IL-1β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease
title_sort impact of caspase-1/11, -3, -7, or il-1β/il-18 deficiency on rabies virus-induced macrophage cell death and onset of disease
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5339016/
https://www.ncbi.nlm.nih.gov/pubmed/28280602
http://dx.doi.org/10.1038/cddiscovery.2017.12
work_keys_str_mv AT kipe impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT nazef impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT suinv impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT vandenberghet impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT francarta impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT lamorals impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT vandenabeelep impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT beyaertr impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT vanguchts impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease
AT kalaim impactofcaspase11137oril1bil18deficiencyonrabiesvirusinducedmacrophagecelldeathandonsetofdisease