1. Academic Validation
  2. ERO1α-dependent endoplasmic reticulum-mitochondrial calcium flux contributes to ER stress and mitochondrial permeabilization by procaspase-activating compound-1 (PAC-1)

ERO1α-dependent endoplasmic reticulum-mitochondrial calcium flux contributes to ER stress and mitochondrial permeabilization by procaspase-activating compound-1 (PAC-1)

  • Cell Death Dis. 2013 Dec 19;4(12):e968. doi: 10.1038/cddis.2013.502.
M Seervi 1 P K Sobhan 1 J Joseph 1 K Ann Mathew 1 T R Santhoshkumar 1
Affiliations

Affiliation

  • 1 Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
Abstract

Procaspase-activating compound-1 (PAC-1) is the first direct caspase-activating compound discovered; using an in vitro cell-free system of Caspase activation. Subsequently, this compound was shown to induce Apoptosis in a variety of Cancer cells with promising in vivo antitumor activity in canine lymphoma model. Recently, we have reported its ability to kill drug-resistant, Bcl-2/Bcl-xL overexpressing and Bax/Bak-deficient cells despite the essential requirement of mitochondrial cytochrome c (cyt. c) release for Caspase activation, indicating that the key molecular targets of PAC-1 in Cancer cells are yet to be identified. Here, we have identified Ero1α-dependent endoplasmic reticulum (ER) calcium leakage to mitochondria through mitochondria-associated ER membranes (MAM) and ER luminal hyper-oxidation as the critical events of PAC-1-mediated cell death. PAC-1 treatment upregulated Ero1α in multiple cell lines, whereas silencing of Ero1α significantly inhibited calcium release from ER and cell death. Loss of ER calcium and hyper-oxidation of ER lumen by Ero1α collectively triggered ER stress. Upregulation of GRP78 and splicing of X-box-binding protein 1 (XBP1) mRNA in multiple Cancer cells suggested ER stress as the general event triggered by PAC-1. XBP1 mRNA splicing and GRP78 upregulation confirmed ER stress even in Bax/Bak double knockout and PAC-1-resistant Apaf-1-knockout cells, indicating an induction of ER stress-mediated mitochondrial Apoptosis by PAC-1. Furthermore, we identified BH3-only protein p53 upregulated modulator of Apoptosis (PUMA) as the key molecular link that orchestrates overwhelmed ER stress to mitochondria-mediated Apoptosis, involving mitochondrial Reactive Oxygen Species, in a p53-independent manner. Silencing of PUMA in Cancer cells effectively reduced cyt. c release and cell death by PAC-1.

Figures
Products