CXCR3 signaling reduces the severity of experimental autoimmune encephalomyelitis by controlling the parenchymal distribution of effector and regulatory T cells in …

M Müller, SL Carter, MJ Hofer, P Manders… - The Journal of …, 2007 - journals.aai.org
M Müller, SL Carter, MJ Hofer, P Manders, DR Getts, MT Getts, A Dreykluft, B Lu, C Gerard…
The Journal of Immunology, 2007journals.aai.org
The chemokine receptor CXCR3 promotes the trafficking of activated T and NK cells in
response to three ligands, CXCL9, CXCL10, and CXCL11. Although these chemokines are
produced in the CNS in multiple sclerosis and experimental autoimmune encephalomyelitis
(EAE), their role in the pathogenesis of CNS autoimmunity is unresolved. We examined the
function of CXCR3 signaling in EAE using mice that were deficient for CXCR3 (CXCR3−/−).
The time to onset and peak disease severity were similar for CXCR3−/− and wild-type (WT) …
Abstract
The chemokine receptor CXCR3 promotes the trafficking of activated T and NK cells in response to three ligands, CXCL9, CXCL10, and CXCL11. Although these chemokines are produced in the CNS in multiple sclerosis and experimental autoimmune encephalomyelitis (EAE), their role in the pathogenesis of CNS autoimmunity is unresolved. We examined the function of CXCR3 signaling in EAE using mice that were deficient for CXCR3 (CXCR3−/−). The time to onset and peak disease severity were similar for CXCR3−/− and wild-type (WT) animals; however, CXCR3−/− mice had more severe chronic disease with increased demyelination and axonal damage. The inflammatory lesions in WT mice consisted of well-demarcated perivascular mononuclear cell infiltrates, mainly in the spinal cord and cerebellum. In CXCR3−/− mice, these lesions were more widespread throughout the CNS and were diffused and poorly organized, with T cells and highly activated microglia/macrophages scattered throughout the white matter. Although the number of CD4+ and CD8+ T cells infiltrating the CNS were similar in CXCR3−/− and WT mice, Foxp3+ regulatory T cells were significantly reduced in number and dispersed in CXCR3−/− mice. The expression of various chemokine and cytokine genes in the CNS was similar in CXCR3−/− and WT mice. The genes for the CXCR3 ligands were expressed predominantly in and/or immediately surrounding the mononuclear cell infiltrates. We conclude that in EAE, CXCR3 signaling constrains T cells to the perivascular space in the CNS and augments regulatory T cell recruitment and effector T cell interaction, thus limiting autoimmune-mediated tissue damage.
journals.aai.org