Your browser doesn't support javascript.
loading
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.
Affiliation
  • Derakhshani S; Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany.
  • Kurz A; Department for Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany.
  • Japtok L; Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.
  • Schumacher F; Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.
  • Pilgram L; Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany.
  • Steinke M; Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany.
  • Kleuser B; Fraunhofer Institute for Silicate Research ISC, Chair of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, University of Wuerzburg, Wuerzburg, Germany.
  • Sauer M; Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.
  • Schneider-Schaulies S; Department for Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany.
  • Avota E; Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany.
Front Immunol ; 10: 1294, 2019.
Article de En | MEDLINE | ID: mdl-31231395
ABSTRACT
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.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Cellules dendritiques / Mouvement cellulaire / Muqueuse respiratoire / Rougeole / Virus de la rougeole Type d'étude: Prognostic_studies Limites: Humans Langue: En Journal: Front Immunol Année: 2019 Type de document: Article Pays d'affiliation: Allemagne

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Cellules dendritiques / Mouvement cellulaire / Muqueuse respiratoire / Rougeole / Virus de la rougeole Type d'étude: Prognostic_studies Limites: Humans Langue: En Journal: Front Immunol Année: 2019 Type de document: Article Pays d'affiliation: Allemagne