Expression and regulation of Fas and Fas ligand on thyrocytes and infiltrating cells during induction and resolution of granulomatous experimental autoimmune …

Y Wei, K Chen, GC Sharp, H Yagita… - The Journal of …, 2001 - journals.aai.org
Y Wei, K Chen, GC Sharp, H Yagita, H Braley-Mullen
The Journal of Immunology, 2001journals.aai.org
Granulomatous experimental autoimmune thyroiditis (G-EAT) is induced by mouse
thyroglobulin-sensitized spleen cells activated in vitro with mouse thyroglobulin, anti-IL-2R,
and IL-12. G-EAT lesions reach maximal severity 19–21 days after cell transfer, and lesions
almost completely resolve by day 35. Depletion of CD8+ cells delays resolution and reduces
Fas ligand (FasL) mRNA expression in thyroids. This study was undertaken to analyze Fas
and FasL protein expression in the thyroid during induction and resolution of G-EAT and to …
Abstract
Granulomatous experimental autoimmune thyroiditis (G-EAT) is induced by mouse thyroglobulin-sensitized spleen cells activated in vitro with mouse thyroglobulin, anti-IL-2R, and IL-12. G-EAT lesions reach maximal severity 19–21 days after cell transfer, and lesions almost completely resolve by day 35. Depletion of CD8+ cells delays resolution and reduces Fas ligand (FasL) mRNA expression in thyroids. This study was undertaken to analyze Fas and FasL protein expression in the thyroid during induction and resolution of G-EAT and to determine whether CD8+ cells might regulate Fas or FasL expression in the thyroid. Fas and FasL expression was analyzed by immunohistochemical staining or in situ hybridization in thyroids of mice with or without depletion of CD8+ cells. Fas and FasL proteins were not detectable in normal thyroids, but expression of both proteins increased during development of G-EAT. Fas was expressed primarily by inflammatory cells; some enlarged thyrocytes were also Fas+. Thyrocytes had intense FasL immunoreactvity, and many CD8+ cells were also FasL positive. Depletion of CD8+ cells resulted in decreased FasL expression by thyrocytes and inflammatory cells, but had no effect on Fas expression. TUNEL assay detected many apoptotic inflammatory cells in proximity to thyrocytes. CD8-depleted thyroids had ongoing inflammation with fewer apoptotic infiltrating cells at day 35. Administration of a neutralizing anti-FasL mAb had no apparent effects on development of G-EAT, but anti-FasL was as effective as anti-CD8 in preventing G-EAT resolution. These results suggested that CD8+ T cells and thyrocytes may kill inflammatory cells through the Fas pathway, contributing to G-EAT resolution.
journals.aai.org