Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature.

Standard

Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature. / Kleinschnitz, Christoph; Kraft, Peter; Dreykluft, Angela; Hagedorn, Ina; Göbel, Kerstin; Schuhmann, Michael K; Langhauser, Friederike; Helluy, Xavier; Schwarz, Tobias; Bittner, Stefan; Mayer, Christian T; Brede, Marc; Varallyay, Csanad; Pham, Mirko; Bendszus, Martin; Jakob, Peter; Magnus, Tim; Meuth, Sven G; Iwakura, Yoichiro; Zernecke, Alma; Sparwasser, Tim; Nieswandt, Bernhard; Stoll, Guido; Wiendl, Heinz.

in: BLOOD, Jahrgang 121, Nr. 4, 4, 2013, S. 679-691.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Kleinschnitz, C, Kraft, P, Dreykluft, A, Hagedorn, I, Göbel, K, Schuhmann, MK, Langhauser, F, Helluy, X, Schwarz, T, Bittner, S, Mayer, CT, Brede, M, Varallyay, C, Pham, M, Bendszus, M, Jakob, P, Magnus, T, Meuth, SG, Iwakura, Y, Zernecke, A, Sparwasser, T, Nieswandt, B, Stoll, G & Wiendl, H 2013, 'Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature.', BLOOD, Jg. 121, Nr. 4, 4, S. 679-691. https://doi.org/10.1182/blood-2012-04-426734

APA

Kleinschnitz, C., Kraft, P., Dreykluft, A., Hagedorn, I., Göbel, K., Schuhmann, M. K., Langhauser, F., Helluy, X., Schwarz, T., Bittner, S., Mayer, C. T., Brede, M., Varallyay, C., Pham, M., Bendszus, M., Jakob, P., Magnus, T., Meuth, S. G., Iwakura, Y., ... Wiendl, H. (2013). Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature. BLOOD, 121(4), 679-691. [4]. https://doi.org/10.1182/blood-2012-04-426734

Vancouver

Bibtex

@article{c87a140ac84b41b4a6d60abbf7828ba2,
title = "Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature.",
abstract = "We have recently identified T cells as important mediators of ischemic brain damage, but the contribution of the different T-cell subsets is unclear. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) are generally regarded as prototypic anti-inflammatory cells that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. In the present study, we examined the role of Tregs after experimental brain ischemia/reperfusion injury. Selective depletion of Tregs in the DEREG mouse model dramatically reduced infarct size and improved neurologic function 24 hours after stroke and this protective effect was preserved at later stages of infarct development. The specificity of this detrimental Treg effect was confirmed by adoptive transfer experiments in wild-type mice and in Rag1(-/-) mice lacking lymphocytes. Mechanistically, Tregs induced microvascular dysfunction in vivo by increased interaction with the ischemic brain endothelium via the LFA-1/ICAM-1 pathway and platelets and these findings were confirmed in vitro. Ablation of Tregs reduced microvascular thrombus formation and improved cerebral reperfusion on stroke, as revealed by ultra-high-field magnetic resonance imaging at 17.6 Tesla. In contrast, established immunoregulatory characteristics of Tregs had no functional relevance. We define herein a novel and unexpected role of Tregs in a primary nonimmunologic disease state.",
keywords = "Animals, Male, Disease Models, Animal, Mice, Mice, Knockout, Cell Communication, Lymphocyte Depletion, Adoptive Transfer, Blood Platelets/immunology/metabolism, Brain/immunology/metabolism/pathology, Brain Ischemia/genetics/*immunology/therapy, Endothelial Cells/immunology/metabolism, Microvessels/pathology/*physiopathology, Stroke/immunology/*metabolism/therapy, T-Lymphocytes, Regulatory/immunology/*metabolism, Animals, Male, Disease Models, Animal, Mice, Mice, Knockout, Cell Communication, Lymphocyte Depletion, Adoptive Transfer, Blood Platelets/immunology/metabolism, Brain/immunology/metabolism/pathology, Brain Ischemia/genetics/*immunology/therapy, Endothelial Cells/immunology/metabolism, Microvessels/pathology/*physiopathology, Stroke/immunology/*metabolism/therapy, T-Lymphocytes, Regulatory/immunology/*metabolism",
author = "Christoph Kleinschnitz and Peter Kraft and Angela Dreykluft and Ina Hagedorn and Kerstin G{\"o}bel and Schuhmann, {Michael K} and Friederike Langhauser and Xavier Helluy and Tobias Schwarz and Stefan Bittner and Mayer, {Christian T} and Marc Brede and Csanad Varallyay and Mirko Pham and Martin Bendszus and Peter Jakob and Tim Magnus and Meuth, {Sven G} and Yoichiro Iwakura and Alma Zernecke and Tim Sparwasser and Bernhard Nieswandt and Guido Stoll and Heinz Wiendl",
year = "2013",
doi = "10.1182/blood-2012-04-426734",
language = "English",
volume = "121",
pages = "679--691",
journal = "BLOOD",
issn = "0006-4971",
publisher = "American Society of Hematology",
number = "4",

}

RIS

TY - JOUR

T1 - Regulatory T cells are strong promoters of acute ischemic stroke in mice by inducing dysfunction of the cerebral microvasculature.

AU - Kleinschnitz, Christoph

AU - Kraft, Peter

AU - Dreykluft, Angela

AU - Hagedorn, Ina

AU - Göbel, Kerstin

AU - Schuhmann, Michael K

AU - Langhauser, Friederike

AU - Helluy, Xavier

AU - Schwarz, Tobias

AU - Bittner, Stefan

AU - Mayer, Christian T

AU - Brede, Marc

AU - Varallyay, Csanad

AU - Pham, Mirko

AU - Bendszus, Martin

AU - Jakob, Peter

AU - Magnus, Tim

AU - Meuth, Sven G

AU - Iwakura, Yoichiro

AU - Zernecke, Alma

AU - Sparwasser, Tim

AU - Nieswandt, Bernhard

AU - Stoll, Guido

AU - Wiendl, Heinz

PY - 2013

Y1 - 2013

N2 - We have recently identified T cells as important mediators of ischemic brain damage, but the contribution of the different T-cell subsets is unclear. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) are generally regarded as prototypic anti-inflammatory cells that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. In the present study, we examined the role of Tregs after experimental brain ischemia/reperfusion injury. Selective depletion of Tregs in the DEREG mouse model dramatically reduced infarct size and improved neurologic function 24 hours after stroke and this protective effect was preserved at later stages of infarct development. The specificity of this detrimental Treg effect was confirmed by adoptive transfer experiments in wild-type mice and in Rag1(-/-) mice lacking lymphocytes. Mechanistically, Tregs induced microvascular dysfunction in vivo by increased interaction with the ischemic brain endothelium via the LFA-1/ICAM-1 pathway and platelets and these findings were confirmed in vitro. Ablation of Tregs reduced microvascular thrombus formation and improved cerebral reperfusion on stroke, as revealed by ultra-high-field magnetic resonance imaging at 17.6 Tesla. In contrast, established immunoregulatory characteristics of Tregs had no functional relevance. We define herein a novel and unexpected role of Tregs in a primary nonimmunologic disease state.

AB - We have recently identified T cells as important mediators of ischemic brain damage, but the contribution of the different T-cell subsets is unclear. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) are generally regarded as prototypic anti-inflammatory cells that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. In the present study, we examined the role of Tregs after experimental brain ischemia/reperfusion injury. Selective depletion of Tregs in the DEREG mouse model dramatically reduced infarct size and improved neurologic function 24 hours after stroke and this protective effect was preserved at later stages of infarct development. The specificity of this detrimental Treg effect was confirmed by adoptive transfer experiments in wild-type mice and in Rag1(-/-) mice lacking lymphocytes. Mechanistically, Tregs induced microvascular dysfunction in vivo by increased interaction with the ischemic brain endothelium via the LFA-1/ICAM-1 pathway and platelets and these findings were confirmed in vitro. Ablation of Tregs reduced microvascular thrombus formation and improved cerebral reperfusion on stroke, as revealed by ultra-high-field magnetic resonance imaging at 17.6 Tesla. In contrast, established immunoregulatory characteristics of Tregs had no functional relevance. We define herein a novel and unexpected role of Tregs in a primary nonimmunologic disease state.

KW - Animals

KW - Male

KW - Disease Models, Animal

KW - Mice

KW - Mice, Knockout

KW - Cell Communication

KW - Lymphocyte Depletion

KW - Adoptive Transfer

KW - Blood Platelets/immunology/metabolism

KW - Brain/immunology/metabolism/pathology

KW - Brain Ischemia/genetics/immunology/therapy

KW - Endothelial Cells/immunology/metabolism

KW - Microvessels/pathology/physiopathology

KW - Stroke/immunology/metabolism/therapy

KW - T-Lymphocytes, Regulatory/immunology/metabolism

KW - Animals

KW - Male

KW - Disease Models, Animal

KW - Mice

KW - Mice, Knockout

KW - Cell Communication

KW - Lymphocyte Depletion

KW - Adoptive Transfer

KW - Blood Platelets/immunology/metabolism

KW - Brain/immunology/metabolism/pathology

KW - Brain Ischemia/genetics/immunology/therapy

KW - Endothelial Cells/immunology/metabolism

KW - Microvessels/pathology/physiopathology

KW - Stroke/immunology/metabolism/therapy

KW - T-Lymphocytes, Regulatory/immunology/metabolism

U2 - 10.1182/blood-2012-04-426734

DO - 10.1182/blood-2012-04-426734

M3 - SCORING: Journal article

C2 - 23160472

VL - 121

SP - 679

EP - 691

JO - BLOOD

JF - BLOOD

SN - 0006-4971

IS - 4

M1 - 4

ER -