[HTML][HTML] Dysregulated autophagy contributes to podocyte damage in Fabry's disease

MC Liebau, F Braun, K Höpker, C Weitbrecht, V Bartels… - PloS one, 2013 - journals.plos.org
MC Liebau, F Braun, K Höpker, C Weitbrecht, V Bartels, RU Müller, S Brodesser, MA Saleem…
PloS one, 2013journals.plos.org
Fabry's disease results from an inborn error of glycosphingolipid metabolism that is due to
deficiency of the lysosomal hydrolase α-galactosidase A. This X-linked defect results in the
accumulation of enzyme substrates with terminally α-glycosidically bound galactose, mainly
the neutral glycosphingolipid Globotriaosylceramide (Gb3) in various tissues, including the
kidneys. Although end-stage renal disease is one of the most common causes of death in
hemizygous males with Fabry's disease, the pathophysiology leading to proteinuria …
Fabry’s disease results from an inborn error of glycosphingolipid metabolism that is due to deficiency of the lysosomal hydrolase α-galactosidase A. This X-linked defect results in the accumulation of enzyme substrates with terminally α-glycosidically bound galactose, mainly the neutral glycosphingolipid Globotriaosylceramide (Gb3) in various tissues, including the kidneys. Although end-stage renal disease is one of the most common causes of death in hemizygous males with Fabry’s disease, the pathophysiology leading to proteinuria, hematuria, hypertension, and kidney failure is not well understood. Histological studies suggest that the accumulation of Gb3 in podocytes plays an important role in the pathogenesis of glomerular damage. However, due to the lack of appropriate animal or cellular models, podocyte damage in Fabry’s disease could not be directly studied yet. As murine models are insufficient, a human model is needed. Here, we developed a human podocyte model of Fabry’s disease by combining RNA interference technology with lentiviral transduction of human podocytes. Knockdown of α-galactosidase A expression resulted in diminished enzymatic activity and slowly progressive accumulation of intracellular Gb3. Interestingly, these changes were accompanied by an increase in autophagosomes as indicated by an increased abundance of LC3-II and a loss of mTOR kinase activity, a negative regulator of the autophagic machinery. These data suggest that dysregulated autophagy in α-galactosidase A-deficient podocytes may be the result of deficient mTOR kinase activity. This finding links the lysosomal enzymatic defect in Fabry’s disease to deregulated autophagy pathways and provides a promising new direction for further studies on the pathomechanism of glomerular injury in Fabry patients.
PLOS