Cargando…

Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium

Transmission of measles virus (MV) from dendritic to airway epithelial cells is considered as crucial to viral spread late in infection. Therefore, pathways and effectors governing this process are promising targets for intervention. To identify these, we established a 3D respiratory tract model whe...

Descripción completa

Detalles Bibliográficos
Autores principales: Derakhshani, Shaghayegh, Kurz, Andreas, Japtok, Lukasz, Schumacher, Fabian, Pilgram, Lisa, Steinke, Maria, Kleuser, Burkhard, Sauer, Markus, Schneider-Schaulies, Sibylle, Avota, Elita
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6560165/
https://www.ncbi.nlm.nih.gov/pubmed/31231395
http://dx.doi.org/10.3389/fimmu.2019.01294
_version_ 1783425915745730560
author Derakhshani, Shaghayegh
Kurz, Andreas
Japtok, Lukasz
Schumacher, Fabian
Pilgram, Lisa
Steinke, Maria
Kleuser, Burkhard
Sauer, Markus
Schneider-Schaulies, Sibylle
Avota, Elita
author_facet Derakhshani, Shaghayegh
Kurz, Andreas
Japtok, Lukasz
Schumacher, Fabian
Pilgram, Lisa
Steinke, Maria
Kleuser, Burkhard
Sauer, Markus
Schneider-Schaulies, Sibylle
Avota, Elita
author_sort Derakhshani, Shaghayegh
collection PubMed
description Transmission of measles virus (MV) from dendritic to airway epithelial cells is considered as crucial to viral spread late in infection. Therefore, pathways and effectors governing this process are promising targets for intervention. To identify these, we established a 3D respiratory tract model where MV transmission by infected dendritic cells (DCs) relied on the presence of nectin-4 on H358 lung epithelial cells. Access to recipient cells is an important prerequisite for transmission, and we therefore analyzed migration of MV-exposed DC cultures within the model. Surprisingly, enhanced motility toward the epithelial layer was observed for MV-infected DCs as compared to their uninfected siblings. This occurred independently of factors released from H358 cells indicating that MV infection triggered cytoskeletal remodeling associated with DC polarization enforced velocity. Accordingly, the latter was also observed for MV-infected DCs in collagen matrices and was particularly sensitive to ROCK inhibition indicating infected DCs preferentially employed the amoeboid migration mode. This was also implicated by loss of podosomes and reduced filopodial activity both of which were retained in MV-exposed uninfected DCs. Evidently, sphingosine kinase (SphK) and sphingosine-1-phosphate (S1P) as produced in response to virus-infection in DCs contributed to enhanced velocity because this was abrogated upon inhibition of sphingosine kinase activity. These findings indicate that MV infection promotes a push-and-squeeze fast amoeboid migration mode via the SphK/S1P system characterized by loss of filopodia and podosome dissolution. Consequently, this enables rapid trafficking of virus toward epithelial cells during viral exit.
format Online
Article
Text
id pubmed-6560165
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-65601652019-06-21 Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium Derakhshani, Shaghayegh Kurz, Andreas Japtok, Lukasz Schumacher, Fabian Pilgram, Lisa Steinke, Maria Kleuser, Burkhard Sauer, Markus Schneider-Schaulies, Sibylle Avota, Elita Front Immunol Immunology Transmission of measles virus (MV) from dendritic to airway epithelial cells is considered as crucial to viral spread late in infection. Therefore, pathways and effectors governing this process are promising targets for intervention. To identify these, we established a 3D respiratory tract model where MV transmission by infected dendritic cells (DCs) relied on the presence of nectin-4 on H358 lung epithelial cells. Access to recipient cells is an important prerequisite for transmission, and we therefore analyzed migration of MV-exposed DC cultures within the model. Surprisingly, enhanced motility toward the epithelial layer was observed for MV-infected DCs as compared to their uninfected siblings. This occurred independently of factors released from H358 cells indicating that MV infection triggered cytoskeletal remodeling associated with DC polarization enforced velocity. Accordingly, the latter was also observed for MV-infected DCs in collagen matrices and was particularly sensitive to ROCK inhibition indicating infected DCs preferentially employed the amoeboid migration mode. This was also implicated by loss of podosomes and reduced filopodial activity both of which were retained in MV-exposed uninfected DCs. Evidently, sphingosine kinase (SphK) and sphingosine-1-phosphate (S1P) as produced in response to virus-infection in DCs contributed to enhanced velocity because this was abrogated upon inhibition of sphingosine kinase activity. These findings indicate that MV infection promotes a push-and-squeeze fast amoeboid migration mode via the SphK/S1P system characterized by loss of filopodia and podosome dissolution. Consequently, this enables rapid trafficking of virus toward epithelial cells during viral exit. Frontiers Media S.A. 2019-06-05 /pmc/articles/PMC6560165/ /pubmed/31231395 http://dx.doi.org/10.3389/fimmu.2019.01294 Text en Copyright © 2019 Derakhshani, Kurz, Japtok, Schumacher, Pilgram, Steinke, Kleuser, Sauer, Schneider-Schaulies and Avota. http://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 Immunology
Derakhshani, Shaghayegh
Kurz, Andreas
Japtok, Lukasz
Schumacher, Fabian
Pilgram, Lisa
Steinke, Maria
Kleuser, Burkhard
Sauer, Markus
Schneider-Schaulies, Sibylle
Avota, Elita
Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium
title Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium
title_full Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium
title_fullStr Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium
title_full_unstemmed Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium
title_short Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium
title_sort measles virus infection fosters dendritic cell motility in a 3d environment to enhance transmission to target cells in the respiratory epithelium
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6560165/
https://www.ncbi.nlm.nih.gov/pubmed/31231395
http://dx.doi.org/10.3389/fimmu.2019.01294
work_keys_str_mv AT derakhshanishaghayegh measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT kurzandreas measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT japtoklukasz measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT schumacherfabian measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT pilgramlisa measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT steinkemaria measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT kleuserburkhard measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT sauermarkus measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT schneiderschauliessibylle measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium
AT avotaelita measlesvirusinfectionfostersdendriticcellmotilityina3denvironmenttoenhancetransmissiontotargetcellsintherespiratoryepithelium