Your browser doesn't support javascript.
loading
ß1-Integrin- and KV1.3 channel-dependent signaling stimulates glutamate release from Th17 cells.
Birkner, Katharina; Wasser, Beatrice; Ruck, Tobias; Thalman, Carine; Luchtman, Dirk; Pape, Katrin; Schmaul, Samantha; Bitar, Lynn; Krämer-Albers, Eva-Maria; Stroh, Albrecht; Meuth, Sven G; Zipp, Frauke; Bittner, Stefan.
Afiliação
  • Birkner K; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Wasser B; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Ruck T; Department of Neurology, University of Muenster, Muenster, Germany.
  • Thalman C; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Luchtman D; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Pape K; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Schmaul S; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Bitar L; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Krämer-Albers EM; Institute for Molecular Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany.
  • Stroh A; Institute for Pathophysiology, FTN, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Meuth SG; Department of Neurology, University of Muenster, Muenster, Germany.
  • Zipp F; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
  • Bittner S; Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn2, ), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
J Clin Invest ; 130(2): 715-732, 2020 02 03.
Article em En | MEDLINE | ID: mdl-31661467
ABSTRACT
Although the impact of Th17 cells on autoimmunity is undisputable, their pathogenic effector mechanism is still enigmatic. We discovered soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) complex proteins in Th17 cells that enable a vesicular glutamate release pathway that induces local intracytoplasmic calcium release and subsequent damage in neurons. This pathway is glutamine dependent and triggered by binding of ß1-integrin to vascular cell adhesion molecule 1 (VCAM-1) on neurons in the inflammatory context. Glutamate secretion could be blocked by inhibiting either glutaminase or KV1.3 channels, which are known to be linked to integrin expression and highly expressed on stimulated T cells. Although KV1.3 is not expressed in CNS tissue, intrathecal administration of a KV1.3 channel blocker or a glutaminase inhibitor ameliorated disability in experimental neuroinflammation. In humans, T cells from patients with multiple sclerosis secreted higher levels of glutamate, and cerebrospinal fluid glutamine levels were increased. Altogether, our findings demonstrate that ß1-integrin- and KV1.3 channel-dependent signaling stimulates glutamate release from Th17 cells upon direct cell-cell contact between Th17 cells and neurons.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Integrina beta1 / Canal de Potássio Kv1.3 / Células Th17 / Esclerose Múltipla Limite: Animals / Humans Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Integrina beta1 / Canal de Potássio Kv1.3 / Células Th17 / Esclerose Múltipla Limite: Animals / Humans Idioma: En Ano de publicação: 2020 Tipo de documento: Article