[HTML][HTML] Autophagic-lysosomal inhibition compromises ubiquitin-proteasome system performance in a p62 dependent manner in cardiomyocytes

Z Tian, C Wang, C Hu, Y Tian, J Liu, X Wang - PloS one, 2014 - journals.plos.org
Z Tian, C Wang, C Hu, Y Tian, J Liu, X Wang
PloS one, 2014journals.plos.org
Intracellular protein degradation is primarily performed by the ubiquitin-proteasome system
(UPS) and the autophagic-lysosomal pathway (ALP). The interplay between these two
pathways has been rarely examined in intact animals and the mechanism underlying the
interplay remains unclear. Hence, we sought to test in vivo and in vitro the impact of
inhibition of the ALP on UPS proteolytic performance in cardiomyocytes and to explore the
underlying mechanism. Transgenic mice ubiquitously expressing a surrogate UPS substrate …
Intracellular protein degradation is primarily performed by the ubiquitin-proteasome system (UPS) and the autophagic-lysosomal pathway (ALP). The interplay between these two pathways has been rarely examined in intact animals and the mechanism underlying the interplay remains unclear. Hence, we sought to test in vivo and in vitro the impact of inhibition of the ALP on UPS proteolytic performance in cardiomyocytes and to explore the underlying mechanism. Transgenic mice ubiquitously expressing a surrogate UPS substrate (GFPdgn) were treated with bafilomycin-A1 (BFA) to inhibit the ALP. Myocardial and renal GFPdgn protein levels but not mRNA levels were increased at 24 hours but not 3 hours after the first injection of BFA. Myocardial protein abundance of key proteasome subunits and the activities of proteasomal peptidases were not discernibly altered by the treatment. In cultured neonatal rat ventricular myocytes (NRVMs), the surrogate UPS substrate GFPu and a control red fluorescence protein (RFP) were co-expressed to probe UPS performance. At 12 hours or 24 hours after ALP inhibition by 3-methyladenine (3-MA) or BFA, GFPu/RFP protein ratios and the protein half-life of GFPu were significantly increased, which is accompanied by increases in p62 proteins. Similar findings were obtained when ALP was inhibited genetically via silencing Atg7 or Rab7. ALP inhibition-induced increases in GFPu and p62 are co-localized in NRVMs. siRNA-mediated p62 knockdown prevented ALP inhibition from inducing GFPu accumulation in NRVMs. We conclude that in a p62-dependent fashion, ALP inhibition impairs cardiac UPS proteolytic performance in cardiomyocytes in vitro and in vivo.
PLOS