PT - JOURNAL ARTICLE AU - Britta Hoechsmann AU - Yoshiko Murakami AU - Makiko Osato AU - Alexej Knaus AU - Michi Kawamoto AU - Norimitsu Inoue AU - Tetsuya Hirata AU - Shogo Murata AU - Markus Anliker AU - Thomas Eggerman AU - Marten Jaeger AU - Ricarda Floettmann AU - Alexander Hoellein AU - Sho Murase AU - Yasutaka Ueda AU - Jun-ichi Nishimura AU - Yuzuru Kanakura AU - Nobuo Kohara AU - Hubert Schrezenmeier AU - Peter M. Krawitz AU - Taroh Kinoshita TI - Complement- and inflammasome-mediated autoinflammation-paroxysmal nocturnal hemoglobinuria AID - 10.1101/635573 DP - 2019 Jan 01 TA - bioRxiv PG - 635573 4099 - http://biorxiv.org/content/early/2019/05/13/635573.short 4100 - http://biorxiv.org/content/early/2019/05/13/635573.full AB - Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired hematopoietic stem cell disorder characterized by complement-mediated hemolysis and thrombosis, and bone marrow failure. Affected cells harbor somatic mutation in X-linked PIGA gene, essential for the initial step in glycosylphosphatidylinositol (GPI) biosynthesis. Loss of GPI biosynthesis results in defective cell-surface expression of GPI-anchored complement regulators CD59 and DAF. The affected stem cells generate many abnormal blood cells after clonal expansion, which occurs under bone marrow failure. Here, we report the mechanistic basis of a disease entity, autoinflammation-paroxysmal nocturnal hemoglobinuria (AIF-PNH), caused by germline mutation plus somatic loss of PIGT on chromosome 20q. A region containing maternally imprinted genes implicated in clonal expansion in 20q-myeloproliferative syndromes was lost together with normal PIGT from paternal chromosome 20. Taking these findings together with a lack of bone marrow failure, the mechanisms of clonal expansion in AIF-PNH appear to differ from those in PNH. AIF-PNH is characterized by intravascular hemolysis and recurrent autoinflammation, such as urticaria, arthralgia, fever and aseptic meningitis. Consistent with PIGT’s essential role in synthesized GPI’s attachment to precursor proteins, non-protein-linked free GPIs appeared on the surface of PIGT-defective cells. PIGT-defective THP-1 cells accumulated higher levels of C3 fragments and C5b-9 complexes, and secreted more IL-1β than PIGA-defective cells after activation of the complement alternative pathway. IL-1β secretion was dependent upon C5b-9 complexes, accounting for the effectiveness of the anti-C5 drug eculizumab for both intravascular hemolysis and autoinflammation. These results suggest that free GPIs enhance complement activation and inflammasome-mediated IL-1β secretion.