RETRACTED: Thapsigargin triggers cardiac contractile dysfunction via NADPH oxidase-mediated mitochondrial dysfunction: Role of Akt dephosphorylation

Free Radic Biol Med. 2011 Dec 15;51(12):2172-2184. doi: 10.1016/j.freeradbiomed.2011.09.005. Epub 2011 Sep 16.

Abstract

ER stress triggers myocardial contractile dysfunction although the underlying mechanism is still elusive. Given that NADPH oxidase was recently implicated in ER stress-induced tissue injury, this study was designed to examine the role of NADPH oxidase in ER stress-induced cardiac mechanical defects and the impact of Akt activation on ER stress-induced cardiac anomalies. Wild-type and transgenic mice with cardiac-specific overexpression of an active mutant of Akt (MyAkt) were subjected to the ER stress inducer thapsigargin (1 and 3mg/kg, ip, for 48h). Thapsigargin compromised echocardiographic parameters, including elevating LVESD and reducing fractional shortening; suppressed cardiomyocyte contractile function, intracellular Ca(2+) handling, and cell survival; and enhanced carbonyl formation, apoptosis, superoxide production, NADPH oxidase expression, and mitochondrial damage. Interestingly, these anomalies were attenuated or mitigated by chronic Akt activation. Treatment with thapsigargin also dephosphorylated Akt and its downstream signal GSK3β (leading to activation of GSK3β), the effect of which was abrogated in MyAkt hearts. Knockdown of the cytosolic subunit of NADPH oxidase, p47(phox), using siRNA abrogated thapsigargin-induced apoptosis and cell death in H9C2 myoblasts. In vitro exposure to thapsigargin induced murine cardiomyocyte dysfunction reminiscent of the in vivo setting, the effects of which were ablated by the NADPH oxidase inhibitor apocynin and the mitochondrial Ca(2+) uptake inhibitor Ru360. In addition, apocynin abrogated thapsigargin-induced loss of mitochondrial membrane potential and permeability transition pore opening, similar to chronic Akt activation. In summary, these data suggest that ER stress interrupts cardiac contractile and intracellular Ca(2+) homeostasis, cell survival, and mitochondrial integrity through an Akt dephosphorylation- and NADPH oxidase-dependent mechanism.

Publication types

  • Research Support, N.I.H., Extramural
  • Retracted Publication

MeSH terms

  • Animals
  • Apoptosis / drug effects
  • Calcium / metabolism
  • Cell Survival / drug effects
  • Cells, Cultured
  • Disease Models, Animal
  • Endoplasmic Reticulum Stress / drug effects
  • Male
  • Membrane Potential, Mitochondrial / drug effects
  • Mice
  • Mice, Transgenic
  • Mitochondria / drug effects*
  • Mitochondria / enzymology
  • Mitochondria / metabolism
  • Mitochondria / pathology
  • Myocardial Contraction / drug effects*
  • Myocytes, Cardiac / metabolism*
  • Myocytes, Cardiac / pathology
  • NADPH Oxidases / metabolism*
  • Phosphorylation / drug effects
  • Proto-Oncogene Proteins c-akt / genetics
  • Proto-Oncogene Proteins c-akt / metabolism*
  • Rats
  • Superoxides / metabolism
  • Thapsigargin / pharmacology*

Substances

  • Superoxides
  • Thapsigargin
  • NADPH Oxidases
  • Proto-Oncogene Proteins c-akt
  • Calcium