The circulating proteinase inhibitor α-1 antitrypsin regulates neutrophil degranulation and autoimmunity

DA Bergin, EP Reeves, K Hurley, R Wolfe… - Science translational …, 2014 - science.org
DA Bergin, EP Reeves, K Hurley, R Wolfe, R Jameel, S Fitzgerald, NG McElvaney
Science translational medicine, 2014science.org
Pathological inflammation and autoimmune disease frequently involve elevated neutrophil
activity in the absence of infectious agents. Tumor necrosis factor–α (TNF-α) contributes to
many of the problems associated with autoimmune diseases. We investigated the ability of
serum α-1 antitrypsin (AAT) to control TNF-α biosynthesis and signaling in neutrophils and
assessed whether AAT deficiency (AATD) is a TNF-α–related disease. In vitro studies
demonstrate that serum AAT coordinates TNF-α intracellular signaling and neutrophil …
Pathological inflammation and autoimmune disease frequently involve elevated neutrophil activity in the absence of infectious agents. Tumor necrosis factor–α (TNF-α) contributes to many of the problems associated with autoimmune diseases. We investigated the ability of serum α-1 antitrypsin (AAT) to control TNF-α biosynthesis and signaling in neutrophils and assessed whether AAT deficiency (AATD) is a TNF-α–related disease. In vitro studies demonstrate that serum AAT coordinates TNF-α intracellular signaling and neutrophil degranulation of tertiary and secondary granules via modulation of ligand-receptor interactions. AATD patients homozygous for the Z allele were characterized by increased activation of the TNF-α system, as demonstrated by increased membrane TNF-α levels and increased plasma concentrations of TNF receptor 1 and neutrophil-released secondary and tertiary granule proteins. The incidence of autoantibodies directed against degranulated lactoferrin and surface protein accessible to these antibodies was increased in ZZ-AATD, leading to an enhanced rate of neutrophil reactive oxygen species production. Treatment of ZZ-AATD individuals with AAT augmentation therapy resulted in decreased membrane TNF-α expression and plasma levels of granule antigenic proteins and immunoglobulin G class autoantibodies. These results provide a mechanism by which AAT augmentation therapy affects TNF-α signaling in the circulating neutrophil, indicating promising potential of this therapy for other TNF-α–related diseases.
AAAS