[HTML][HTML] Uromodulin p. Cys147Trp mutation drives kidney disease by activating ER stress and apoptosis

BG Johnson, LT Dang, G Marsh… - The Journal of …, 2017 - Am Soc Clin Investig
BG Johnson, LT Dang, G Marsh, AM Roach, ZG Levine, A Monti, D Reyon, L Feigenbaum…
The Journal of clinical investigation, 2017Am Soc Clin Investig
Uromodulin-associated kidney disease (UAKD) is caused by mutations in the uromodulin
(UMOD) gene that result in a misfolded form of UMOD protein, which is normally secreted by
nephrons. In UAKD patients, mutant UMOD is poorly secreted and accumulates in the ER of
distal kidney epithelium, but its role in disease progression is largely unknown. Here, we
modeled UMOD accumulation in mice by expressing the murine equivalent of the human
UMOD p. Cys148Trp point mutation (Umod C147W/+ mice). Like affected humans, these …
Uromodulin-associated kidney disease (UAKD) is caused by mutations in the uromodulin (UMOD) gene that result in a misfolded form of UMOD protein, which is normally secreted by nephrons. In UAKD patients, mutant UMOD is poorly secreted and accumulates in the ER of distal kidney epithelium, but its role in disease progression is largely unknown. Here, we modeled UMOD accumulation in mice by expressing the murine equivalent of the human UMOD p.Cys148Trp point mutation (UmodC147W/+ mice). Like affected humans, these UmodC147W/+ mice developed spontaneous and progressive kidney disease with organ failure over 24 weeks. Analysis of diseased kidneys and purified UMOD-producing cells revealed early activation of the PKR-like ER kinase/activating transcription factor 4 (PERK/ATF4) ER stress pathway, innate immune mediators, and increased apoptotic signaling, including caspase-3 activation. Unexpectedly, we also detected autophagy deficiency. Human cells expressing UMOD p.Cys147Trp recapitulated the findings in UmodC147W/+ mice, and autophagy activation with mTOR inhibitors stimulated the intracellular removal of aggregated mutant UMOD. Human cells producing mutant UMOD were susceptible to TNF-α– and TRAIL-mediated apoptosis due to increased expression of the ER stress mediator tribbles-3. Blocking TNF-α in vivo with the soluble recombinant fusion protein TNFR:Fc slowed disease progression in UmodC147W/+ mice by reducing active caspase-3, thereby preventing tubule cell death and loss of epithelial function. These findings reveal a targetable mechanism for disease processes involved in UAKD.
The Journal of Clinical Investigation