Abstract

The execution phase of apoptosis is a critical process in programmed cell death in response to a multitude of cellular stresses. A crucial component of this pathway is the apoptosome, a platform for the activation of pro-caspase 9 (PC9). Recent findings have shown that autocleavage of PC9 to Caspase 9 (C9) p35/p12 not only permits XIAP-mediated C9 inhibition but also temporally shuts down apoptosome activity, forming a molecular timer. In order to delineate the combined contributions of XIAP and the apoptosome molecular timer to apoptosis execution we utilised a systems modelling approach. We demonstrate that cooperative recruitment of PC9 to the apoptosome, based on existing PC9-apoptosome interaction data, is important for efficient formation of PC9 homodimers, autocatalytic cleavage and dual regulation by XIAP and the molecular timer across biologically relevant PC9 and APAF1 concentrations. Screening physiologically relevant concentration ranges of apoptotic proteins, we discovered that the molecular timer can prevent apoptosis execution in specific scenarios after complete or partial mitochondrial outer membrane permeabilisation (MOMP). Furthermore, its ability to prevent apoptosis is intricately tied to a synergistic combination with XIAP. Finally, we demonstrate that simulations of these processes are prognostic of survival in stage III colorectal cancer and that the molecular timer may promote apoptosis resistance in a subset of patients. Based on our findings, we postulate that the physiological function of the molecular timer is to aid XIAP in the shutdown of caspase-mediated apoptosis execution. This shutdown potentially facilitates switching to pro-inflammatory caspase-independent responses subsequent to Bax/Bak pore formation.

Highlights

  • Apoptosis is the process of controlled cell death, integral for development and maintaining homoeostasis

  • We demonstrated that homodimerisation-based activation of Pro-Caspase 9 (PC9) was insufficient to explain experimental data on subsequent caspase 3 (C3)-activity and we suggested that allosteric activation likely contributes to PC9 activation at the apoptosome [15]

  • Overexpression of X-linked inhibitor of apoptosis protein (XIAP) failed to notably decrease the level of cleaved caspase 3 demonstrating poor inhibition of apoptosome activity (Fig. 1d). This suggested that PC9 autocleavage, a necessary step for both the molecular timer and XIAP inhibition, was inadequate and the observed C3-substrate cleavage was mainly driven by PC9 monomers

Read more

Summary

Introduction

Apoptosis is the process of controlled cell death, integral for development and maintaining homoeostasis. Apoptosis signalling is strictly controlled by a family of cysteine aspartate specific proteases (Caspases), which are expressed as inactive zymogens (ProCaspases) [2]. The execution phase of apoptosis is triggered in response to multiple extrinsic and intrinsic stress signals by Bax/Bak-mediated mitochondrial outer membrane permeabilisation (MOMP) [3]. MOMP allows the release of the mitochondrial proteins cytochrome c (CytC) and second mitochondrial-derived activator of caspases (SMAC) into the cytosol, initiating the apoptosis execution phase cascade [3]. CytC interacts with apoptotic peptidase activating factor 1 (APAF1) and along with ATP causes its oligomerisation into a heptameric complex called the apoptosome. Pro-Caspase 9 (PC9) is recruited to the apoptosome platform, which is

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.