Abstract

Poly(ADP-Ribose) polymerases (PARPs) are enzymes that metabolize NAD+. PARP1 and PARP10 were previously implicated in the regulation of autophagy. Here we showed that cytosolic electron-dense particles appear in the cytoplasm of C2C12 myoblasts in which PARP2 is silenced by shRNA. The cytosolic electron-dense bodies resemble autophagic vesicles and, in line with that, we observed an increased number of LC3-positive and Lysotracker-stained vesicles. Silencing of PARP2 did not influence the maximal number of LC3-positive vesicles seen upon chloroquine treatment or serum starvation, suggesting that the absence of PARP2 inhibits autophagic breakdown. Silencing of PARP2 inhibited the activity of AMP-activated kinase (AMPK) and the mammalian target of rapamycin complex 2 (mTORC2). Treatment of PARP2-silenced C2C12 cells with AICAR, an AMPK activator, nicotinamide-riboside (an NAD+ precursor), or EX-527 (a SIRT1 inhibitor) decreased the number of LC3-positive vesicles cells to similar levels as in control (scPARP2) cells, suggesting that these pathways inhibit autophagic flux upon PARP2 silencing. We observed a similar increase in the number of LC3 vesicles in primary PARP2 knockout murine embryonic fibroblasts. We provided evidence that the enzymatic activity of PARP2 is important in regulating autophagy. Finally, we showed that the silencing of PARP2 induces myoblast differentiation. Taken together, PARP2 is a positive regulator of autophagic breakdown in mammalian transformed cells and its absence blocks the progression of autophagy.

Highlights

  • Poly(ADP-ribose) (PAR) metabolism is an evolutionarily conserved posttranslational modification of proteins [1]

  • We assessed the function of a set of energy sensors in scPARP2 and shPARP2 cells and we found a deregulation of cellular energy sensors. mTORC1 activity, measured through assessing the phosphorylation of the p70 S6 kinase (Phospho-p70 S6 Kinase (Thr389)), did not change in the shPARP2 cells (Figure 6A)

  • We showed that the genetic or pharmacological silencing of PARP2 induces the number of autophagic vesicles in cellular models through inhibiting AMPK and inducing SIRT1 activity

Read more

Summary

Introduction

Poly(ADP-ribose) (PAR) metabolism is an evolutionarily conserved posttranslational modification of proteins [1]. PAR is synthesized by members of the Poly(ADP-Ribose) polymerase enzyme family, among them, PARP1 and PARP2 [2]. PARP2 accounts for 15–20% of total cellular PARP activity [5,6]. Both enzymes are involved in a plethora of cellular processes, among these, in the regulation of cellular energy and metabolic homeostasis [1,7,8,9]. Autophagy is a process in cells that is dedicated to the removal of damaged cellular proteins and components (e.g., mitochondria) [10,11]. In our current study we showed an opposite impact of PARP2 on autophagy as compared to PARP1

Cell Culture
Transient Transfection
Immunofluorescence and Confocal Microscopy
LysoTracker Deep Red Staining
SDS-PAGE and Western Blotting
Statistical Analysis
Silencing of PARP2 Induces Autophagy in C2C12 Cells
Validation ofofPARP2 silencing stably-transfected
Silencing
The Activity of PARP2 Plays Role in Mediating Autophagy
Silencing of PARP2
Discussion
Full Text
Published version (Free)

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