[PDF][PDF] Deficient activity of the nuclease MRE11A induces T cell aging and promotes arthritogenic effector functions in patients with rheumatoid arthritis

Y Li, Y Shen, P Hohensinner, J Ju, Z Wen… - Immunity, 2016 - cell.com
Y Li, Y Shen, P Hohensinner, J Ju, Z Wen, SB Goodman, H Zhang, JJ Goronzy, CM Weyand
Immunity, 2016cell.com
Immune aging manifests with a combination of failing adaptive immunity and insufficiently
restrained inflammation. In patients with rheumatoid arthritis (RA), T cell aging occurs
prematurely, but the mechanisms involved and their contribution to tissue-destructive
inflammation remain unclear. We found that RA CD4+ T cells showed signs of aging during
their primary immune responses and differentiated into tissue-invasive, proinflammatory
effector cells. RA T cells had low expression of the double-strand-break repair nuclease …
Summary
Immune aging manifests with a combination of failing adaptive immunity and insufficiently restrained inflammation. In patients with rheumatoid arthritis (RA), T cell aging occurs prematurely, but the mechanisms involved and their contribution to tissue-destructive inflammation remain unclear. We found that RA CD4+ T cells showed signs of aging during their primary immune responses and differentiated into tissue-invasive, proinflammatory effector cells. RA T cells had low expression of the double-strand-break repair nuclease MRE11A, leading to telomeric damage, juxtacentromeric heterochromatin unraveling, and senescence marker upregulation. Inhibition of MRE11A activity in healthy T cells induced the aging phenotype, whereas MRE11A overexpression in RA T cells reversed it. In human-synovium chimeric mice, MRE11Alow T cells were tissue-invasive and pro-arthritogenic, and MRE11A reconstitution mitigated synovitis. Our findings link premature T cell aging and tissue-invasiveness to telomere deprotection and heterochromatin unpacking, identifying MRE11A as a therapeutic target to combat immune aging and suppress dysregulated tissue inflammation.
cell.com