Elsevier

Experimental Cell Research

Volume 313, Issue 13, 1 August 2007, Pages 2920-2936
Experimental Cell Research

Research Article
Two small enzyme isoforms mediate mammalian mitochondrial poly(ADP-ribose) glycohydrolase (PARG) activity

https://doi.org/10.1016/j.yexcr.2007.03.043Get rights and content

Abstract

Poly(ADP-ribose)glycohydrolase (PARG) is the major enzyme capable of rapidly hydrolyzing poly(ADP-ribose) (PAR) formed by the diverse members of the PARP enzyme family. This study presents an alternative splice mechanism by which two novel PARG protein isoforms of 60 kDa and 55 kDa are expressed from the human PARG gene, termed hPARG60 and hPARG55, respectively. Homologous forms were found in the mouse (mPARG63 and mPARG58) supporting the hypothesis that expression of small PARG isoforms is conserved among mammals. A PARG protein of ∼ 60 kDa has been described for decades but with its genetic basis unknown, it was hypothesized to be a product of posttranslational cleavage of larger PARG isoforms. While this is not excluded entirely, isolation and expression of cDNA clones from different sources of RNA indicate that alternative splicing leads to expression of a catalytically active hPARG60 in multiple cell compartments. A second enzyme, hPARG55, that can be expressed through alternative translation initiation from hPARG60 transcripts is strictly targeted to the mitochondria. Functional studies of a mitochondrial targeting signal (MTS) in PARG exon IV suggest that hPARG60 may be capable of shuttling between nucleus and mitochondria, which would be in line with a proposed function of PAR in genotoxic stress-dependent, nuclear–mitochondrial crosstalk.

Introduction

Poly(ADP-ribosyl)ation is a transient, dynamic and reversible posttranslational modification of proteins involved in a host of biological functions [1], [2]. Poly(ADP-ribose) (PAR) is formed by all PARP enzymes by cleavage of NAD+ into nicotinamide and ADP-ribose which becomes polymerized and transferred to specific target proteins. Genome-wide sequence analyses in human suggest the existence of up to 17 different genes encoding enzymes capable of synthesizing poly(ADP-ribose) (PAR) [3], [4], [5], and, so far, ten of them have been shown to be catalytically active. In contrast, only three human poly(ADP-ribose) glycohydrolase (PARG) proteins, PARG111, PARG102 and PARG99, have been described, which are all expressed from one single PARG gene by alternative splicing [6], [7], [8]. PARG proteins have been the only enzymes known to efficiently catalyze hydrolysis of poly(ADP-ribose) (PAR) produced by enzymes of the poly(ADP-ribose) polymerase (PARP) family [9] until recently a novel gene encoding a 39 kDa enzyme with low poly(ADP-ribose) glycohydrolase activity has been discovered [10], [11], [12]. However, the extent to which this enzyme, (ADP-ribose) hydrolase (ARH3), that is structurally unrelated to PARG, participates in the rapid, DNA damage dependent PAR turnover within the cell has not been elucidated yet.

PARG rapidly cleaves PAR to form monomeric ADP-ribose by both exoglycosidic and endoglycosidic activity. Following DNA damage, PAR metabolism is mediated by PARP-1 and PARP-2 in concert with PARG, and PAR turnover can be extremely rapid with a half-life of the biopolymer of only ∼ 1 min [13]. Cellular accumulation of PAR beyond a short period of time has been shown to be deleterious [14], [15] and is involved in caspase-independent cell death regulation ([16] reviewed in [17]). Alterations of PAR metabolism are causally involved in the pathogenesis of inflammatory [19] and autoimmune disease [20], ischemia–reperfusion injury in brain [21], heart and intestine [22], [23], [24], neuronal degeneration and neurotoxicity [25], genetic and genomic instability (reviewed in [26]) and cancer (reviewed in [27], [28]). Targeting of the poly(ADP-ribose) metabolic pathway has therefore become an important focus in research on cancer therapy (recent reviews in [29], [30], [31]), cardiovascular disease intervention [32], [33], [34], [35] and a number of other pathophysiological conditions [36], [37]. Because of the potential of PAR metabolism as a drug target, identification of the key players appears to be crucial. While several PARP enzymes have been studied extensively, comparatively little is known about regulation of the catabolic arm of the pathway. However, the function of PARG in the PAR pathway is essential as deletion of the PARG gene results in a chronic, lethally toxic accumulation of cellular PAR [14], [38].

In contrast, a hypomorphic PARGΔ2–3/Δ2–3 mutant mouse that is devoid of exons II and III [18] was shown to be viable and expressing a residual PARG of 63 kDa by alternative splicing of the disrupted gene. This protein, designated mPARG63, is catalytically active and has unique properties [18]. In the present study we tested the hypothesis that the residual PARG proteins in the hypomorphic mouse may in fact be proteins that are naturally also expressed in wildtype mice as well as human. We show that mPARG63 is a naturally occurring protein that is expressed not only in the hypomorphic mutant but also in wildtype mouse tissues. More significantly, we demonstrate the existence of a human homolog, hPARG60, that is expressed in all tissues tested (skin, liver, testis, HeLa). At mRNA level, human PARG60 (hPARG60) is characterized as missing exon V, making it slightly smaller in size than the mouse homolog mPARG63. The data presented support the hypothesis that alternative translation initiation allows for expression of a protein with a consensus mitochondrial targeting sequence of the presequence type with strictly mitochondrial localization, termed hPARG55 (mPARG58 in mouse). The identification of hPARG60 (mPARG63) and hPARG55 (mPARG58) as novel PARG isoforms that associate with the mitochondria is of particular interest with regard to the emerging view that ADP-ribose metabolism may be involved in mitochondria-mediated, caspase-independent cell death pathways of biomedical interest [15], [39].

Section snippets

Cloning of human PARG cDNA by 5′RACE and construction of expression vectors

Human total RNA preparations from skin, liver and testis were purchased from Stratagene (La Jolla, CA). Mouse total RNA from liver and testis was also purchased or isolated from PARGΔ2–3/Δ2–3 mouse embryonic fibroblast culture, reversely transcribed and used in subsequent polymerase chain reaction (PCR) amplification reactions as described earlier [6], [7]. A mouse or human PARG gene specific primer which binds in the distal portion of the PARG 5′-UTR (human: hex0for:

Isolation of a cDNA encoding a novel hPARG60 isoform

PCR amplification of hPARG cDNA (Fig. 1A) from reversely transcribed total RNA yielded numerous bands rather than a single product. In order to amplify specifically the 5′ ends of cDNA derived from PARG transcripts, a forward primer in the 5′ untranslated region (hex0for) in combination with a reverse primer in exon VI (hex6rev) was used (5′UTR, Fig. 1A, left hand side of the gel). These primers produced three bands corresponding to the cDNAs of hPARG111 (band 1, 1880 bp), hPARG102 (band 2,

Discussion

The results of the study presented here suggest the presence of an additional level of complexity of cellular PAR metabolism due to the catalytic activity of novel small PARG isoforms associated with the mitochondria. This view is supported by several conclusions that can be drawn from this study: (i) two novel protein isoforms, hPARG60 and hPARG55, were discovered which are capable of PAR hydrolysis. They are conserved from mouse to human and are ubiquitously expressed by alternative splicing

Acknowledgments

We thank Donna Coyle for preparing 32P-labeled PAR and Zhao-Qi Wang, Universitaet Jena, for providing PARGΔ2–3/Δ2–3 3T3 cells. This work was supported by NIH grants CA-43894 (MKJ) and HD048837 (RGM).

References (69)

  • A. Chiarugi

    Poly(ADP-ribosyl)ation and stroke

    Pharmacol. Res.

    (2005)
  • P.A. Nguewa et al.

    Poly(ADP-ribose) polymerases: homology, structural domains and functions. Novel therapeutical applications

    Prog. Biophys. Mol. Biol.

    (2005)
  • E. Winstall et al.

    Preferential perinuclear localization of poly(ADP-ribose) glycohydrolase

    Exp. Cell Res.

    (1999)
  • C.C. Kiehlbauch et al.

    High resolution fractionation and characterization of ADP-ribose polymers

    Anal. Biochem.

    (1993)
  • S. Tanuma et al.

    Identification of two activities of (ADP-ribose)n glycohydrolase in HeLa S3 cells

    Biochem. Biophys. Res. Commun.

    (1986)
  • S. Tanuma et al.

    Occurrence of (ADP-ribose)N glycohydrolase in human erythrocytes

    Biochem. Biophys. Res. Commun.

    (1986)
  • K. Uchida et al.

    Preferential degradation of protein-bound (ADP-ribose)n by nuclear poly(ADP-ribose) glycohydrolase from human placenta

    J. Biol. Chem.

    (1993)
  • W. Lin et al.

    Isolation and characterization of the cDNA encoding bovine poly(ADP-ribose) glycohydrolase

    J. Biol. Chem.

    (1997)
  • C. Keil et al.

    MNNG-induced cell death is controlled by interactions between PARP-1, poly (ADP-ribose) glycohydrolase and XRCC1

    J. Biol. Chem.

    (2006)
  • H. Gao et al.

    Altered poly(ADP-ribose) metabolism impairs cellular responses to genotoxic stress in a hypomorphic mutant of poly(ADP-ribose) glycohydrolase

    Exp. Cell Res.

    (2007)
  • K.N. Truscott et al.

    Mechanisms of protein import into mitochondria

    Curr. Biol.

    (2003)
  • P.O. Hassa et al.

    Nuclear ADP-ribosylation reactions in mammalian cells: where are we today and where are we going?

    Microbiol. Mol. Biol. Rev.

    (2006)
  • D. D'Amours et al.

    Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions

    Biochem. J.

    (1999)
  • V. Schreiber et al.

    Poly(ADP-ribose): novel functions for an old molecule

    Nat. Rev., Mol. Cell Biol.

    (2006)
  • R.G. Meyer et al.

    Enzymes in poly(ADP-ribose) metabolism

  • S. Kernstock et al.

    Cloning, expression, purification, crystallization and preliminary X-ray diffraction analysis of human ARH3, the first eukaryotic protein-ADP-ribosylhydrolase

    Acta Crystallogr. Sect. F. Struct. Biol. Cryst. Commun.

    (2006)
  • C. Mueller-Dieckmann et al.

    The structure of human ADP-ribosylhydrolase 3 (ARH3) provides insights into the reversibility of protein ADP-ribosylation

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • H. Juarez-Salinas et al.

    Poly(ADP-ribose) levels in carcinogen-treated cells

    Nature

    (1979)
  • D.W. Koh et al.

    Failure to degrade poly(ADP-ribose) causes increased sensitivity to cytotoxicity and early embryonic lethality

    Proc. Natl. Acad. Sci. U. S. A.

    (2004)
  • S.W. Yu et al.

    Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor

    Science

    (2002)
  • C.G. Nevill-Manning, C.N. Huang, D.L. Brutlag, Pairwise protein sequence alignment using Needleman-Wunsch and...
  • U. Cortes et al.

    Depletion of the 110-kilodalton isoform of poly(ADP-ribose) glycohydrolase increases sensitivity to genotoxic and endotoxic stress in mice

    Mol. Cell. Biol.

    (2004)
  • M. Masutani et al.

    Poly(ADP-ribosyl)ation in relation to cancer and autoimmune disease

    Cell. Mol. Life Sci.

    (2005)
  • A. Cozzi et al.

    Poly(ADP-ribose) accumulation and enhancement of postischemic brain damage in 110-kDa poly(ADP-ribose) glycohydrolase null mice

    J. Cereb. Blood Flow Metab.

    (2005)
  • Cited by (66)

    • Poly (ADP-ribose) (PAR)-dependent cell death in neurodegenerative diseases

      2020, International Review of Cell and Molecular Biology
      Citation Excerpt :

      PARG-catalyzed hydrolysis of PAR polymers yields free ADP-ribose units through its endoglycosidic and exoglycosidic activity (O'Sullivan et al., 2019). PARG exists in alternatively spliced isoforms with full-length PARG (110 kDa) localized in the nucleus and the splice variants in diverse localization patterns (Bonicalzi et al., 2003; Haince et al., 2006; Meyer et al., 2007; Meyer-Ficca et al., 2004). Many studies have demonstrated the importance of PARG in parthanatos.

    • Niacin

      2018, Advances in Food and Nutrition Research
      Citation Excerpt :

      Through alternative splicing, a single PARG gene gives rise to several PARG protein isoforms of different sizes. Importantly, the different subcellular localization allows PARG to degrade PAR molecules present in different parts of the cell (Cortes et al., 2004; Koh et al., 2004; Meyer, Meyer-Ficca, Whatcott, Jacobson, & Jacobson, 2007; Meyer-Ficca, Meyer, Coyle, Jacobson, & Jacobson, 2004; Winstall et al., 1999). The terminal ADP-ribose unit at the receptor glutamate amino acid residue in a PAR-modified target protein can be removed by MACROD1 and MACROD2 proteins, and by TARG1/C6ord130 (Chen et al., 2011; Sharifi et al., 2013).

    • PARPing for balance in the homeostasis of poly(ADP-ribosyl)ation

      2017, Seminars in Cell and Developmental Biology
    View all citing articles on Scopus

    This work was supported by NIH grants CA-43894 (MKJ) and HD048837 (RGM).

    View full text