Show simple item record

dc.contributor.authorDöring, Marius
dc.contributor.authorBlees, Hanna
dc.contributor.authorKoller, Nicole
dc.contributor.authorTischer-Zimmermann, Sabine
dc.contributor.authorMüsken, Mathias
dc.contributor.authorHenrich, Frederik
dc.contributor.authorBecker, Jennifer
dc.contributor.authorGrabski, Elena
dc.contributor.authorWang, Junxi
dc.contributor.authorJanssen, Hans
dc.contributor.authorZuschratter, Werner
dc.contributor.authorNeefjes, Jacques
dc.contributor.authorKlawonn, Frank
dc.contributor.authorEiz-Vesper, Britta
dc.contributor.authorTampé, Robert
dc.contributor.authorKalinke, Ulrich
dc.date.accessioned2019-04-29T13:56:09Z
dc.date.available2019-04-29T13:56:09Z
dc.date.issued2019-03-26
dc.identifier.citationBlood Adv. 2019 Mar 26;3(6):839-850. doi: 10.1182/bloodadvances.2018027268.en_US
dc.identifier.issn2473-9537
dc.identifier.pmid30867143
dc.identifier.doi10.1182/bloodadvances.2018027268
dc.identifier.urihttp://hdl.handle.net/10033/621757
dc.description.abstractDendritic cells (DCs) take up antigen in the periphery, migrate to secondary lymphoid organs, and present processed antigen fragments to adaptive immune cells and thus prime antigen-specific immunity. During local inflammation, recirculating monocytes are recruited from blood to the inflamed tissue, where they differentiate to macrophages and DCs. In this study, we found that monocytes showed high transporter associated with antigen processing (TAP)–dependent peptide compartmentalization and that after antigen pulsing, they were not able to efficiently stimulate antigen-specific T lymphocytes. Nevertheless, upon in vitro differentiation to monocyte-derived DCs, TAP-dependent peptide compartmentalization as well as surface major histocompatibility complex I turnover decreased and the cells efficiently restimulated T lymphocytes. Although TAP-dependent peptide compartmentalization decreased during DC differentiation, TAP expression levels increased. Furthermore, TAP relocated from early endosomes in monocytes to the endoplasmic reticulum (ER) and lysosomal compartments in DCs. Collectively, these data are compatible with the model that during monocyte-to-DC differentiation, the subcellular relocation of TAP and the regulation of its activity assure spatiotemporal separation of local antigen uptake and processing by monocytes and efficient T-lymphocyte stimulation by DCs.en_US
dc.language.isoenen_US
dc.publisherAmerican Society of Hematologyen_US
dc.rightsAttribution-NonCommercial-ShareAlike 4.0 International*
dc.rights.urihttp://creativecommons.org/licenses/by-nc-sa/4.0/*
dc.titleModulation of TAP-dependent antigen compartmentalization during human monocyte-to-DC differentiation.en_US
dc.typeArticleen_US
dc.contributor.departmentTWINCORE, Zentrum für experimentelle und klinische Infektionsforschung GmbH, Feodor-Lynen-Str.7,30625 Hannover, Germany.en_US
dc.identifier.journalBlood Advancesen_US
refterms.dateFOA2019-04-29T13:56:10Z
dc.source.journaltitleBlood advances


Files in this item

Thumbnail
Name:
Döring et al.pdf
Size:
1.544Mb
Format:
PDF
Description:
Open Access publication

This item appears in the following Collection(s)

Show simple item record

Attribution-NonCommercial-ShareAlike 4.0 International
Except where otherwise noted, this item's license is described as Attribution-NonCommercial-ShareAlike 4.0 International