Cargando…

CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration

Dendritic cells (DCs) are potent and versatile professional antigen-presenting cells and central for the induction of adaptive immunity. The ability to migrate and transport peripherally acquired antigens to draining lymph nodes for subsequent cognate T cell priming is a key feature of DCs. Conseque...

Descripción completa

Detalles Bibliográficos
Autores principales: Uetz-von Allmen, Edith, Samson, Guerric P. B., Purvanov, Vladimir, Maeda, Takahiro, Legler, Daniel F.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8482423/
https://www.ncbi.nlm.nih.gov/pubmed/34603281
http://dx.doi.org/10.3389/fimmu.2021.702453
_version_ 1784576899470589952
author Uetz-von Allmen, Edith
Samson, Guerric P. B.
Purvanov, Vladimir
Maeda, Takahiro
Legler, Daniel F.
author_facet Uetz-von Allmen, Edith
Samson, Guerric P. B.
Purvanov, Vladimir
Maeda, Takahiro
Legler, Daniel F.
author_sort Uetz-von Allmen, Edith
collection PubMed
description Dendritic cells (DCs) are potent and versatile professional antigen-presenting cells and central for the induction of adaptive immunity. The ability to migrate and transport peripherally acquired antigens to draining lymph nodes for subsequent cognate T cell priming is a key feature of DCs. Consequently, DC-based immunotherapies are used to elicit tumor-antigen specific T cell responses in cancer patients. Understanding chemokine-guided DC migration is critical to explore DCs as cellular vaccines for immunotherapeutic approaches. Currently, research is hampered by the lack of appropriate human cellular model systems to effectively study spatio-temporal signaling and CCR7-driven migration of human DCs. Here, we report that the previously established human neoplastic cell line CAL-1 expresses the human DC surface antigens CD11c and HLA-DR together with co-stimulatory molecules. Importantly, if exposed for three days to GM-CSF, CAL-1 cells induce the endogenous expression of the chemokine receptor CCR7 upon encountering the clinically approved TLR7/8 agonist Resiquimod R848 and readily migrate along chemokine gradients. Further, we demonstrate that CAL-1 cells can be genetically modified to express fluorescent (GFP)-tagged reporter proteins to study and visualize signaling or can be gene-edited using CRISPR/Cas9. Hence, we herein present the human CAL-1 cell line as versatile and valuable cellular model system to effectively study human DC migration and signaling.
format Online
Article
Text
id pubmed-8482423
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-84824232021-10-01 CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration Uetz-von Allmen, Edith Samson, Guerric P. B. Purvanov, Vladimir Maeda, Takahiro Legler, Daniel F. Front Immunol Immunology Dendritic cells (DCs) are potent and versatile professional antigen-presenting cells and central for the induction of adaptive immunity. The ability to migrate and transport peripherally acquired antigens to draining lymph nodes for subsequent cognate T cell priming is a key feature of DCs. Consequently, DC-based immunotherapies are used to elicit tumor-antigen specific T cell responses in cancer patients. Understanding chemokine-guided DC migration is critical to explore DCs as cellular vaccines for immunotherapeutic approaches. Currently, research is hampered by the lack of appropriate human cellular model systems to effectively study spatio-temporal signaling and CCR7-driven migration of human DCs. Here, we report that the previously established human neoplastic cell line CAL-1 expresses the human DC surface antigens CD11c and HLA-DR together with co-stimulatory molecules. Importantly, if exposed for three days to GM-CSF, CAL-1 cells induce the endogenous expression of the chemokine receptor CCR7 upon encountering the clinically approved TLR7/8 agonist Resiquimod R848 and readily migrate along chemokine gradients. Further, we demonstrate that CAL-1 cells can be genetically modified to express fluorescent (GFP)-tagged reporter proteins to study and visualize signaling or can be gene-edited using CRISPR/Cas9. Hence, we herein present the human CAL-1 cell line as versatile and valuable cellular model system to effectively study human DC migration and signaling. Frontiers Media S.A. 2021-09-16 /pmc/articles/PMC8482423/ /pubmed/34603281 http://dx.doi.org/10.3389/fimmu.2021.702453 Text en Copyright © 2021 Uetz-von Allmen, Samson, Purvanov, Maeda and Legler https://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
Uetz-von Allmen, Edith
Samson, Guerric P. B.
Purvanov, Vladimir
Maeda, Takahiro
Legler, Daniel F.
CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration
title CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration
title_full CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration
title_fullStr CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration
title_full_unstemmed CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration
title_short CAL-1 as Cellular Model System to Study CCR7-Guided Human Dendritic Cell Migration
title_sort cal-1 as cellular model system to study ccr7-guided human dendritic cell migration
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8482423/
https://www.ncbi.nlm.nih.gov/pubmed/34603281
http://dx.doi.org/10.3389/fimmu.2021.702453
work_keys_str_mv AT uetzvonallmenedith cal1ascellularmodelsystemtostudyccr7guidedhumandendriticcellmigration
AT samsonguerricpb cal1ascellularmodelsystemtostudyccr7guidedhumandendriticcellmigration
AT purvanovvladimir cal1ascellularmodelsystemtostudyccr7guidedhumandendriticcellmigration
AT maedatakahiro cal1ascellularmodelsystemtostudyccr7guidedhumandendriticcellmigration
AT leglerdanielf cal1ascellularmodelsystemtostudyccr7guidedhumandendriticcellmigration