Cargando…

Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke

Rag1(−/−) mice, lacking functional B and T cells, have been extensively used as an adoptive transfer model to evaluate neuroinflammation in stroke research. However, it remains unknown whether natural killer (NK) cell development and functions are altered in Rag1(−/−) mice as well. This connection h...

Descripción completa

Detalles Bibliográficos
Autores principales: Rolfes, Leoni, Ruck, Tobias, David, Christina, Mencl, Stine, Bock, Stefanie, Schmidt, Mariella, Strecker, Jan-Kolja, Pfeuffer, Steffen, Mecklenbeck, Andreas-Schulte, Gross, Catharina, Gliem, Michael, Minnerup, Jens, Schuhmann, Michael K., Kleinschnitz, Christoph, Meuth, Sven G.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer US 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8766401/
https://www.ncbi.nlm.nih.gov/pubmed/34105078
http://dx.doi.org/10.1007/s12975-021-00923-3
_version_ 1784634521831866368
author Rolfes, Leoni
Ruck, Tobias
David, Christina
Mencl, Stine
Bock, Stefanie
Schmidt, Mariella
Strecker, Jan-Kolja
Pfeuffer, Steffen
Mecklenbeck, Andreas-Schulte
Gross, Catharina
Gliem, Michael
Minnerup, Jens
Schuhmann, Michael K.
Kleinschnitz, Christoph
Meuth, Sven G.
author_facet Rolfes, Leoni
Ruck, Tobias
David, Christina
Mencl, Stine
Bock, Stefanie
Schmidt, Mariella
Strecker, Jan-Kolja
Pfeuffer, Steffen
Mecklenbeck, Andreas-Schulte
Gross, Catharina
Gliem, Michael
Minnerup, Jens
Schuhmann, Michael K.
Kleinschnitz, Christoph
Meuth, Sven G.
author_sort Rolfes, Leoni
collection PubMed
description Rag1(−/−) mice, lacking functional B and T cells, have been extensively used as an adoptive transfer model to evaluate neuroinflammation in stroke research. However, it remains unknown whether natural killer (NK) cell development and functions are altered in Rag1(−/−) mice as well. This connection has been rarely discussed in previous studies but might have important implications for data interpretation. In contrast, the NOD-Rag1(null)IL2rg(null) (NRG) mouse model is devoid of NK cells and might therefore eliminate this potential shortcoming. Here, we compare immune-cell frequencies as well as phenotype and effector functions of NK cells in Rag1(−/−) and wildtype (WT) mice using flow cytometry and functional in vitro assays. Further, we investigate the effect of Rag1(−/−) NK cells in the transient middle cerebral artery occlusion (tMCAO) model using antibody-mediated depletion of NK cells and adoptive transfer to NRG mice in vivo. NK cells in Rag1(−/−) were comparable in number and function to those in WT mice. Rag1(−/−) mice treated with an anti-NK1.1 antibody developed significantly smaller infarctions and improved behavioral scores. Correspondingly, NRG mice supplemented with NK cells were more susceptible to tMCAO, developing infarctions and neurological deficits similar to Rag1(−/−) controls. Our results indicate that NK cells from Rag1(−/−) mice are fully functional and should therefore be considered in the interpretation of immune-cell transfer models in experimental stroke. Fortunately, we identified the NRG mice, as a potentially better-suited transfer model to characterize individual cell subset-mediated neuroinflammation in stroke. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12975-021-00923-3.
format Online
Article
Text
id pubmed-8766401
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Springer US
record_format MEDLINE/PubMed
spelling pubmed-87664012022-02-02 Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke Rolfes, Leoni Ruck, Tobias David, Christina Mencl, Stine Bock, Stefanie Schmidt, Mariella Strecker, Jan-Kolja Pfeuffer, Steffen Mecklenbeck, Andreas-Schulte Gross, Catharina Gliem, Michael Minnerup, Jens Schuhmann, Michael K. Kleinschnitz, Christoph Meuth, Sven G. Transl Stroke Res Original Article Rag1(−/−) mice, lacking functional B and T cells, have been extensively used as an adoptive transfer model to evaluate neuroinflammation in stroke research. However, it remains unknown whether natural killer (NK) cell development and functions are altered in Rag1(−/−) mice as well. This connection has been rarely discussed in previous studies but might have important implications for data interpretation. In contrast, the NOD-Rag1(null)IL2rg(null) (NRG) mouse model is devoid of NK cells and might therefore eliminate this potential shortcoming. Here, we compare immune-cell frequencies as well as phenotype and effector functions of NK cells in Rag1(−/−) and wildtype (WT) mice using flow cytometry and functional in vitro assays. Further, we investigate the effect of Rag1(−/−) NK cells in the transient middle cerebral artery occlusion (tMCAO) model using antibody-mediated depletion of NK cells and adoptive transfer to NRG mice in vivo. NK cells in Rag1(−/−) were comparable in number and function to those in WT mice. Rag1(−/−) mice treated with an anti-NK1.1 antibody developed significantly smaller infarctions and improved behavioral scores. Correspondingly, NRG mice supplemented with NK cells were more susceptible to tMCAO, developing infarctions and neurological deficits similar to Rag1(−/−) controls. Our results indicate that NK cells from Rag1(−/−) mice are fully functional and should therefore be considered in the interpretation of immune-cell transfer models in experimental stroke. Fortunately, we identified the NRG mice, as a potentially better-suited transfer model to characterize individual cell subset-mediated neuroinflammation in stroke. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12975-021-00923-3. Springer US 2021-06-08 2022 /pmc/articles/PMC8766401/ /pubmed/34105078 http://dx.doi.org/10.1007/s12975-021-00923-3 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Original Article
Rolfes, Leoni
Ruck, Tobias
David, Christina
Mencl, Stine
Bock, Stefanie
Schmidt, Mariella
Strecker, Jan-Kolja
Pfeuffer, Steffen
Mecklenbeck, Andreas-Schulte
Gross, Catharina
Gliem, Michael
Minnerup, Jens
Schuhmann, Michael K.
Kleinschnitz, Christoph
Meuth, Sven G.
Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke
title Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke
title_full Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke
title_fullStr Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke
title_full_unstemmed Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke
title_short Natural Killer Cells Are Present in Rag1(−/−) Mice and Promote Tissue Damage During the Acute Phase of Ischemic Stroke
title_sort natural killer cells are present in rag1(−/−) mice and promote tissue damage during the acute phase of ischemic stroke
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8766401/
https://www.ncbi.nlm.nih.gov/pubmed/34105078
http://dx.doi.org/10.1007/s12975-021-00923-3
work_keys_str_mv AT rolfesleoni naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT rucktobias naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT davidchristina naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT menclstine naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT bockstefanie naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT schmidtmariella naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT streckerjankolja naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT pfeuffersteffen naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT mecklenbeckandreasschulte naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT grosscatharina naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT gliemmichael naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT minnerupjens naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT schuhmannmichaelk naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT kleinschnitzchristoph naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke
AT meuthsveng naturalkillercellsarepresentinrag1miceandpromotetissuedamageduringtheacutephaseofischemicstroke