Abstract

BackgroundThe formation of ADP-ribose polymers on target proteins by poly(ADP-ribose) polymerases serves a variety of cell signaling functions. In addition, extensive activation of poly(ADP-ribose) polymerase-1 (PARP-1) is a dominant cause of cell death in ischemia-reperfusion, trauma, and other conditions. Poly(ADP-ribose) glycohydrolase (PARG) degrades the ADP-ribose polymers formed on acceptor proteins by PARP-1 and other PARP family members. PARG exists as multiple isoforms with differing subcellular localizations, but the functional significance of these isoforms is uncertain.Methods / Principal FindingsPrimary mouse astrocytes were treated with an antisense phosphorodiamidate morpholino oligonucleotide (PMO) targeted to exon 1 of full-length PARG to suppress expression of this nuclear-specific PARG isoform. The antisense-treated cells showed down-regulation of both nuclear PARG immunoreactivity and nuclear PARG enzymatic activity, without significant alteration in cytoplasmic PARG activity. When treated with the genotoxic agent MNNG to induced PARP-1 activation, the antisense-treated cells showed a delayed rate of nuclear PAR degradation, reduced nuclear condensation, and reduced cell death.Conclusions/SignificanceThese results support a preferentially nuclear localization for full-length PARG, and suggest a key role for this isoform in the PARP-1 cell death pathway.

Highlights

  • Poly(ADP-ribose) polymerases consume NAD+ to form poly(ADP-ribose) on acceptor proteins

  • In the present study we aimed to examine the specific role of nuclear Poly(ADP-ribose) glycohydrolase (PARG) activity in poly(ADP-ribose) polymerase-1 (PARP-1) – mediated cell death by selectively blocking translation of the 111 kDa, nuclear-targeted isoform using an antisense phosphorodiamidate morpholino oligonucleotide (PMO) targeted to the 59 start of exon 1 (Fig. 1A)

  • Immunostaining for PARG in mouse astrocyte cultures treated with antisense PARG PMO showed a marked and selective loss of immunoreactivity from the nuclei of the antisense PMO-treated cells, as compared to sister cultures treated with control PMO or no PMO (Fig. 1B)

Read more

Summary

Introduction

Poly(ADP-ribose) polymerases consume NAD+ to form poly(ADP-ribose) (abbreviated as PAR) on acceptor proteins. This post-translational modification influences protein-protein interactions and serves a variety of cell signaling functions [1]. Extensive activation of PARP-1 occurs after ischemia-reperfusion, trauma, and other conditions that cause DNA damage. In these settings, PARP-1 activation leads to NAD+ depletion, mitochondrial release of apoptosisinducing factor, and cell death [3,4,5]. Extensive activation of poly(ADP-ribose) polymerase-1 (PARP-1) is a dominant cause of cell death in ischemia-reperfusion, trauma, and other conditions. Poly(ADP-ribose) glycohydrolase (PARG) degrades the ADP-ribose polymers formed on acceptor proteins by PARP-1 and other PARP family members. PARG exists as multiple isoforms with differing subcellular localizations, but the functional significance of these isoforms is uncertain

Objectives
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