Abstract
ADP-ribosylation refers to the addition of one or more ADP-ribose units onto protein substrates and this protein modification has been implicated in various cellular processes including DNA damage repair, RNA metabolism, transcription and cell cycle regulation. This review focuses on a compilation of large-scale proteomics studies that identify ADP-ribosylated proteins and their associated proteins by mass spectrometry using a variety of enrichment strategies. Some methods, such as the use of a poly(ADP-ribose)-specific antibody and boronate affinity chromatography and NAD+ analogues, have been employed for decades while others, such as the use of protein microarrays and recombinant proteins that bind ADP-ribose moieties (such as macrodomains), have only recently been developed. The advantages and disadvantages of each method and whether these methods are specific for identifying mono(ADP-ribosyl)ated and poly(ADP-ribosyl)ated proteins will be discussed. Lastly, since poly(ADP-ribose) is heterogeneous in length, it has been difficult to attain a mass signature associated with the modification sites. Several strategies on how to reduce polymer chain length heterogeneity for site identification will be reviewed.
Keywords: ADP-ribosylation, Mono(ADP-ribose), PARP, Poly(ADP-ribose), Proteomics, Sirtuin
1. Introduction
ADP-ribosylation is a post-translational modification in which one or more ADP-ribose moieties from NAD+ is transferred to a protein substrate [1-8] (Figure 1). If only one ADP-ribose is transferred, it is known as mono(ADP-ribosyl)ation (MARylation). Poly(ADP-ribosyl)ation (PARylation) occurs when additional ADP-ribose moieties are added to the first ADP-ribose. Excellent reviews have extensively covered these modifications regarding their enzymes and cellular pathways involved [1-8]. Here we review the recent development of proteomics techniques to determine the substrate identities and their modification sites, with a focus on technical aspects of enrichment strategies.
1.1 PARylation
PARylation is known to be attached to glutamate, aspartate and lysine residues of proteins by a subclass of ADP-ribosyltransferases, commonly known as poly(ADP-ribose) polymerases (PARPs) [1,2]. PARPs catalyze the initial transfer of ADP-ribose from NAD+ to a target protein as well as the subsequent addition of (up to ~200) ADP-ribose units onto the first one. Recent systematic analyses have shown that 4 out of the 17 human ADP-ribosyltransferases (PARP1, PARP2, PARP5a, PARP5b) exhibit PARylating activities in vitro (Table 1) [9]. Notably, PARP4, when in complex with other proteins, can also add multiple ADP-ribose units inside cells [10]. PARP homologues are found in animals, plants, fungi and protist kingdoms, as well as prokaryotes and viruses [11,12]. In animals and plants, PARylation is implicated in key cellular processes including DNA repair, telomere length maintenance, transcription, post-transcriptional gene regulation, immune responses, and stress responses [1,2,13-15]. Since modification sites were not identified in many studies, it has been difficult to dissect how PARylation mechanistically regulates protein functions. Yet, the physiological importance of PARylation is evident because animal knockout models of PARPs display a range of phenotypes (reviewed in [16]). These include altered inflammatory and stress responses, increased tumor incidence, and developmental and neurological abnormalities. On the other hand, PARP inhibitors in clinical trials have shown promise in treating cancers, neurodegenerative disorders, heart attacks and ischemia [13,17]. Thus, identifying PARylation sites becomes paramount not only to solve long outstanding biological questions on the functions of poly(ADP-ribose) (PAR), but also to translate this biological knowledge to clinical settings.
Table 1.
Synthesis | Degradation | |
---|---|---|
PARylation | PARP1, PARP2, PARP4*, PARP5a, PARP5b | PARG, ARH3 |
MARylation | PARP3, PARP6, PARP7, PARP8, PARP10, PARP11, PARP12, PARP14, PARP15, PARP16, SIRT1, SIRT2, SIRT3, SIRT4, SIRT5, SIRT6, SIRT7, ART1, ART3, ART4, ART5 | MacroD1, MacroD2, C6orf130/TARG1, ARH1 |
Like other protein modifications, the presence of PARylation at a single residue may alter the activities of modified substrates or their interactions with other proteins non-covalently. Interestingly, such non-covalent binding is critically determined by the length and structure of the PAR chain [18-20]. As ADP-ribose subunits are sequentially added to a protein covalently, the emanating PAR chain can be bound by specific proteins non-covalently. As a result, scaffolds of protein-PAR::protein interactions are formed (reviewed in [18]). Such scaffolding property is observed in the recruitment of DNA repair proteins at the site of DNA damage [21], and for the assembly of spindle poles [22] and RNA organelles such as stress granules [14].
PARylation can be reversed mainly through two classes of degradation enzymes – one that can break the ribose-ribose bonds within the PAR chain and the other that breaks the covalent bonds between the proximal ADP-ribose units and the modified proteins. The principal degradation enzyme that breaks the ribose-ribose bonds within PAR is encoded by a singe gene named PAR glycohydrolase (PARG) that has three catalytically active splice isoforms [23-25]. Recent data indicate that another enzyme, ADP-ribosylhydrolase 3 (ARH3), can also cleave PAR in vitro and in cells (particularly in mitochondria) [25-27]. However, PARG cannot cleave the proximal ADP-ribose groups from its modified protein, effectively converting a PARylated protein into a MARylated one [28]. It is currently unclear whether ARH3 can remove proximal ADP-ribose groups from modified proteins. Recently, three enzymes were identified that can remove proximal ADP-ribose groups from modified acidic residues – MacroD1, MacroD2 and TARG1/C6orf130 [29-31]. Such stepwise arrangement in PAR degradation suggests the potential importance of the regulatory transition between PARylation and MARylation.
1.2 MARylation
MARylation was originally identified as an important mechanism of bacterial pathogenesis catalyzed by their secreted toxins including diphtheria, pertussis, cholera and clostridial toxins [3,4]. These toxins permanently modify functions of crucial eukaryotic host cell proteins, such as the α-subunit of G proteins, the small GTPase Rho, monomeric actin and elongation factor 2. Subsequently, MARylation was discovered in mammalian extracellular surface proteins that help mediate immune responses. In contrast, relatively few proteins were identified in humans as intracellular substrates and little is known about which enzymes MARylate intracellular substrates [3,4], even though there could be up to 700 times more MARylated than PARylated residues inside cells [32-34].
Based on structural and bioinformatics analyses, 9 of the 17 ADP-ribosyltransferases were predicted to have MARylating activities [2] (Table 1). Recently, Vyas and colleagues used in vitro biochemical assays to demonstrate that the majority (11) of the 17 PARP family members have MARylating activity, while 4 have PARylating activity, and the remaining 2 members are inactive [9]. Similar activities were also demonstrated in another family of NAD+-consuming enzymes known as sirtuins [35]. This family is exemplified by the yeast member Sir2p, which catalyzes the removal of acetyl groups from histones and is involved in gene silencing, chromosomal stability, and aging. Yet, Sir2p can also catalyze the transfer of ADP-ribose to itself and histones. In humans, all seven human homologues, SIRT1-7, possess MARylating activities in vitro [3]. Two endogenous substrates were identified to date – SIRT4 ribosylates glutamate dehydrogenase to suppress insulin signaling in pancreatic β cells [36] and SIRT6 MARylates PARP1, which allows for its automodification with further ADP-ribose units upon DNA damage [37]. This latter observation suggests that the two NAD+-dependent signaling pathways can crosstalk and cooperate [38], and highlights the possibility that MARylated residues can be PARylated in vivo.
A perplexing observation is that ADP-ribosylation has been identified at amino acids of very different chemistries including cysteine, diphthamide (modified histidine), glutamate, arginine, and lysine [3,4,9,37]. However, only few enzymes involved in the removal of modified residues have been identified – ADP-ribosylhydrolase 1 (ARH1) for ADP-ribosylated arginine, and MacroD1, MacroD2, TARG1 for modified glutamate. Therefore, it is currently not clear whether all classes of MARylation are reversible.
1.3 Challenges for identification of ADP-ribosylation sites by mass spectrometry
There are two primary challenges to identify both classes of ADP-ribosylation sites: (1) the labile bonds between ADP-ribose and protein during peptide fragmentation and (2) the low abundance of the modification. Recent developments in peptide fragmentation techniques and improvements in mass spectrometry sensitivity have partially mitigated these challenges (reviewed in [39]). In Section 2, various enrichment strategies and their potential applications towards identifying MARylated and PARylated substrates will be discussed. For PARylation, an additional challenge is that the modification is heterogeneous in length (2-200 ADP-ribose subunits) and therefore no unique mass signatures can be assigned to this protein modification. Different strategies to tackle this problem will be reviewed in Section 3. Current proteomics studies are summarized in Table 2.
Table 2.
References | Method | # proteins identified | Moiety Identification | Covalent or Non-covalent | PAR induction | PARG inhibitors | PARP inhibitors | |
---|---|---|---|---|---|---|---|---|
Poly | Mono | |||||||
Gagné et al., 2008 | 10H IP, LC-MS/MS | 334 | Yes | No | Both | MNNG | No | No |
Dani et al. 2009 | Af1521 pull down, LC-MS/MS and MALDI MS | 12 | Yes | Yes | Both | No | No | No |
Troiani et al., 2011 | Biotinylated NAD analogue and protein microarray | 51 (PARP2) | Yes | No | Covalent | No | No | No |
Jiang et al. 2011 | Clickable NAD analogue and biotin affinity tag | 79 | Yes | No | Covalent | No | No | No |
Gagné et al., 2012 | 10H IP, Af1521 and PARG-DEAD pull down, GeLC-MS/MS | 665 | Yes | (Yes for Af1521) | Both | MNNG | ADP-HPD | ABT-888 |
Isabelle et al. 2012 | 10H IP, nanoLC-MS/MS (temporal dynamics using SILAC) | 163 | Yes | No | Both | MNNG | ADP-HPD | No |
Jungmichel et al, 2013 | Af1521 pull down, LC-MS/MS | 235 | Yes | Yes | Both | H2O2, IR, MMS, UV | siRNA and ADP-HPD | PJ34 and 3AB |
Feijs et al., 2013 | Biotinylated NAD analogue and protein microarray | 78 (PARP10), 142 (PARP14) | No | Yes | COvalent | No | No | No |
Zhang et al., 2013 | Boronate, Hydroxylamine Elution | 340 | Yes | Yes | Covalent | H2O2, MNNG | shRNA | 3-AB, olaparib, A966942, AG14361, iniparib |
Carter-O'Connell et al., 2014 | Clickable NAD with analogue-sensitive PARP and biotin affinity tag | 42 (PARP1), 301 (PARP2) | No | Yes | Covalent | No | No | No |
Daniels et al., 2014 | Snake Venom Phosphodiesterase, Phosphopeptide enrichment | 21 (human), 33 (mouse) | Yes | Yes | Covalent | MNNG | No | No |
2. ENRICHMENT STRATEGIES
2.1 Antibody-based approaches
2.1.1 PAR-specific 10H antibodies
One standard method to isolate PARylated proteins from cell lysate is the use of a mouse monoclonal antibody 10H developed in 1984 by the Miwa group [40]. The immunogen used to create this antibody was PAR of more than 20 ADP-ribose residues (20mers). The monoclonal antibody was shown through competitive binding assays and protection assays to bind linear portions of several ADP-ribose units [40]. Competitive binding assays showed that antibody binding to PAR was inhibited strongly by >20mers, moderately by 10-30mers, and weakly by the smallest structural unit of PAR, iso-ADP-ribose (c.f. Figure 1C), but not with ADP-ribose, AMP, poly(A), RNA nor DNA [40]. These data indicated that 10H could be used for specifically enriching PARylated proteins but not MARylated proteins. Subsequent studies using surface plasmon resonance detected the binding affinity of 10H to 14-16mers and 55-63mers in the picomolar range [41]. Stoichiometry analyses revealed that up to 21 10H molecules were bound to one 63mer of PAR, suggesting one 10H molecule binds 3-4 ADP-ribose units [41].
Given its strong affinity to PAR, 10H is routinely used to enrich PARylated proteins in immunoprecipitation experiments, including its use in chromatin immunoprecipitation experiments to identify where PAR impacts chromatin structure (e.g. [42]). So far, three large-scale proteomics analyses have been performed using this antibody by the Poirier group [43-45]. They have investigated the genotoxic response upon treatment with alkylating agent N-methyl-N′-nitro-N-nitrosoguanidine (MNNG) in multiple cell lines. Using quantitative proteomics, they mapped how cellular pathways are affected by PAR through its network of associated proteins in a detailed temporal manner [44,45]. Since PAR can both covalently conjugate to proteins and bind proteins avidly in a non-covalent manner, the Poirier group has carefully noted that these studies identified proteins associated with PAR, rather than PARylated substrates [43-45]. This is especially true when considering that the non-covalent PAR-mediated tight association with histones withstands phenol-partitioning, strong acid, detergents, and high salt [46].
Another consideration when using 10H antibodies is that it cannot efficiently capture polymers of 10 ADP-ribose units or less, as reported in ref. [47]. While the 10H antibody is capable of binding heterogeneous PAR lengths [40], the exact epitope of the antibody remains unclear. Therefore, the identification may lead to the exclusion of proteins interacting with or modified by short oligomers of ADP-ribose, or PARylated proteins containing an epitope different than that of 10H.
2.1.2 Other antibodies
Though there are several antibodies commercially available that can recognize PAR by immunoblot and immunofluorescence [14,47], they seem not to be efficient for immunoprecipitation. Similarly, several attempts have been made to develop antibodies for detection of specific MARylated proteins but they are not generally applicable for wider use [48-52]. Therefore, the field will benefit from the development of new antibodies to detect PARylated and MARylated proteins. In particular, the Miwa group previously demonstrated that it is possible to develop monoclonal antibodies against the branched portions of PAR [40]. The development of branch-specific antibodies will be extremely useful to discern the function of this protein-modification with such distinct structures.
2.2 Approaches using biological modules that recognize ADP-ribose
2.2.1 Macrodomains identify MARylated and PARylated proteins
Several protein domains that bind mono- and poly(ADP-ribose) have been identified, such as the WWE domain, PBZ (PAR-binding zinc finger) domain, and PBM (PAR-binding motif) (Table 3; reviewed in ref. [5,6,53]). However, the macrodomain is most commonly observed in the literature as bait in pull-down experiments to isolate ADP-ribosylated proteins from cell lysates in large-scale proteomics studies [45,54,55]. The macrodomain, originally discovered in the histone variant macroH2A [56], possesses a highly conserved structure from bacteria to viruses to eukaryotes [57]. As evident from Table 3, several PARPs contain macrodomains. In addition, sirtuins, the NAD+ consuming enzymes mentioned earlier, are also closely linked to macrodomains, suggesting the intimate link between macrodomains and ADP-ribosylation [58]. The first macrodomain structure solved was from the archaebacteria Archaeoglobus fulgidus, named Af1521, and displays a unique fold made up of a β-sheet surrounded by α-helices [59]. While the structures of macrodomains are highly conserved, the sequences of macrodomains from eukaryotes and viruses vary, which may account for the observed differences in binding specificities. Some macrodomains (such as macroH2A1.1) show binding specificity for PARylated proteins [60,61], while others (such as macrodomains 2 and 3 in PARP14) bind MARylated proteins [62] (Table 3). In addition, some macrodomain-containing proteins possess hydrolase activity, which, as discussed in the subsequent sections, should be taken into consideration when using macrodomain proteins to enrich for ADP-ribosylated proteins.
Table 3.
Name | Binding | Hydrolysis | ||
---|---|---|---|---|
PAR | MAR | PAR | MAR | |
ALC1 macrodomain | Yes | – | – | – |
MacroH2A1.1 macrodomain | Yes | No | No | No |
MacroH2A1.2 macrodomain | No | – | – | – |
MacroH2A2 macrodomain | No | – | – | – |
PARP9 macrodomain | Yes | – | – | – |
PARP14 macrodomain | No | Yes | – | No |
PARP15 macrodomain | – | – | – | – |
MacroD1 | Yes | – | No | Yes |
MacroD2 | No | – | No | Yes |
MacroD3 | No | – | – | No |
C6orf130 | Yes | – | No | Yes |
PARG | – | – | Yes | No |
Af1521 | Yes | Yes | No | Yes |
HUWE1 WWE | Yes | No | – | – |
RNF146 WWE | Yes | No | – | – |
CHFR PBZ | Yes | No | – | – |
APLF PBZ | Yes | No | – | – |
2.2.2 Using the Af1521 macrodomain
Dani and colleagues were the first to use the Af1521 macrodomain to enrich for ADP-ribosylated proteins [54]. Af1521 was chosen because of its ability to recognize free ADP-ribose with high affinities and specificities (Kd = 0.13 μM; >1000 fold greater affinity than NAD+ or adenosine) [63]. As a proof of principle, they first tested whether Af1521 can pull down the αi and β subunits of heterotrimeric G proteins, well-characterized substrates of bacterial toxin mono-ADP-ribosyltransferases modified at cysteine and arginine, respectively [54]. To set up a pipeline for isolating ADP-ribosylated proteins, Dani et al also established two critical experimental conditions for elution and lysate pre-clearing. As Af1521 can bind avidly to free ADP-ribose, the latter is used to specifically elute ADP-ribosylated proteins from the macrodomain. As a negative control, a G42E mutant that is sufficient to abrogate ADP-ribose binding was used [63]. They proposed to first pre-clear the lysate with this site-specific mutant, followed by pull down with the wild-type Af1521 macrodomain; the modified proteins are then eluted with ADP-ribose and analyzed by mass spectrometry. Using this pipeline with MALDI-TOF-MS or LC-MS/MS, they identified 12 proteins, which include known endogenous MARylated and PARylated substrates modified at different amino acids.
Jungmichel and colleagues recently utilized Af1521 aiming to enrich for PARylated proteins after treatment with four different types of DNA damaging agents in U2OS cells [55]. To distinguish non-specific proteins bound to Af1521 non-covalently from ADP-ribosylated proteins, the quantitative proteomic technique SILAC [64] was used to differentially label lysates prepared for pulldown experiments with wild-type and ADP-ribose binding-deficient mutant Af1521 macrodomains. Overall, such strategies revealed an enrichment of 235 proteins. One caveat of this macrodomain enrichment approach is that it cannot distinguish whether the identified substrates are MARylated or PARylated. Given that the pull down experiments were performed under non-denaturing conditions, as in the case of 10H antibody enrichment, it is not possible to definitively distinguish whether the identified proteins are covalently attached to or non-covalently bound to ADP-ribose subunit(s). Of another cautionary note, Af1521 macrodomains have been recently reported to have hydrolase activity that removes ADP-ribose groups from modified acidic residues (glutamate/aspartate) [29,30], this macrodomain-based enrichment approach, therefore, could have potential bias in the identification towards ADP-ribosylated substrates with modified neutral/basic residues. Indeed, Daniels et al recently showed that Af1521 macrodomain enrichment has a distinct bias against glutamate, but not aspartate, residues, as revealed by a newly developed proteomics pipeline to identify endogenous MARylated/PARylated sites [65] (see also Section 3.3). Therefore, a mutant of Af1521 macrodomain that lacks the hydrolase activity but retains its proficiency to bind ADP-ribose could potentially be engineered to avoid the loss of substrates with modified glutamate residues.
2.2.3 Other ADP-ribose binding modules
Gagné et al (2012) proposed a substrate-trapping approach by using a catalytically inactive E756D mutant of PARG (PARG-DEAD approach) [45]. Given that PARG shares structural similarity to the macrodomain [28,66,67], they reasoned that the site-specific mutant can form stable interactions with PAR for affinity purification of PARylated proteins and their associated binding partners [45]. To identify these PAR-associated protein complexes, a GFP-tagged PARG construct encompassing the catalytically inactive mutation was transfected into cells for 24 hours prior to cell lysis and anti-GFP immunoprecipitation. By comparing data from the PARG-DEAD approach to the Af1521 macrodomain and 10H affinity-based purification, they reported significant overlap between datasets: 73% of 10H-enriched proteins and 96% of Af1521 macrodomain-enriched proteins were identified by the PARG DEAD approach, respectively. Such significant overlap suggests the feasibility and validity of this substrate-trapping approach. In addition, the overlap in identified proteins by the PARG-DEAD and Af1521 macrodomain suggests that these protein domains may have similar substrate specificities. One obvious bias, however, is that the PARG-DEAD approach also enriches for PARG-specific interacting proteins. Interestingly, another notable difference between datasets is the bias toward different cellular compartments – the Af1521 macrodomain isolated only nuclear proteins, the PARG-DEAD approach preferentially enriched for nuclear proteins while the 10H preferred mitochondrial proteins. This inherent bias may be a result of the prominent nuclear localization of GFP-PARG, but could also indicate that these different enrichment methods may have an affinity for distinct pools of ADP-ribosylated proteins due to differences in length and structure of the protein modification. In addition to the difference in cellular localization and specific interacting partners, the difference in the number of identified proteins by PARG-DEAD (561) and Af1521 macrodomain (95) could be the result of the possible hydrolase bias, previously discussed. It would therefore be of interest to test whether a hydrolase-dead but substrate-trapping Af1521 macrodomain will enrich for an ADP-ribose interacting partner profile more similar to that identified by the PARG-DEAD approach.
In addition to using the archaebacterial Af1521, isolated macrodomains from human Histone H2A.1 have been used to enrich for ADP-ribosylated proteins, including known substrates PARP1, Ku70/80, and DNA-PKc [61]. Similarly, macrodomains from human PARP14 have been used to pull down MARylated PARP10 [62]. As both of these macrodomains do not have hydrolase activities [29,30], they may serve as alternatives to Af1521 for enrichment of endogenous ADP-ribosylated proteins. Given that structural and biochemical analyses revealed that the binding specificity of these domains are dependent on the neighboring amino acids surrounding the modified sites [62,68], these macrodomains will likely enrich for a restricted set of native ADP-ribosylated partners within cells. Besides macrodomains, the WWE domain from RNF146 has also been shown to be responsible for binding PARylated PARP1 and PARP5a [69-71]. Given that the WWE domain specifically recognizes the smallest structural subunit for PAR, iso-ADP-ribose [72], and that this domain is currently commercially available, it could be a useful addition to specifically enrich for PARylated proteins. As increasingly more biological modules that recognize ADP-ribose are discovered, additional tools will be available for analysis of ADP-ribosylated proteomes.
2.3 Boronate enrichment
Boronate affinity chromatography is a chemical method of isolating PAR by interacting with the cis-diol group of the ribose (c.f. Figure 1A), and has been used for decades to enrich and quantitate the amount of MARylated and PARylated proteins [32-34]. Assuming an average chain length of 10 units for each PAR chain, these studies found that there are up to 700 times more MARylated residues than PARylated residues inside cells. Moreover, the ratio of mono to poly(ADP-ribose) levels varies with development, indicating independent changes of these two classes of protein modifications and distinct functions of MARylated proteins [33]. In the 1970s, boronate resin was also first used to purify ADP-ribosylated proteins from rat liver nuclei and the incorporation of radioactivity with 14C-NAD+ was utilized as a measurement of the retention of ADP-ribosylated proteins [73]. They found that while 95% of proteins, applied with a buffer pH of 8.2, passed through the column, 80% of the radioactivity was retained and eluted at pH 6.0. However, it was also noted in this study that part of the retained radioactivity observed may be free-polymer or monomers released from proteins due to the basic pH conditions (e.g. ADP-ribosylated residues conjugated at acidic residues are particularly susceptible to high pH [74]). Nonetheless, enough ADP-ribosylated proteins were retained for identification: mainly histone proteins H1 and H2B.
Alvarez-Gonzalez and colleagues published an evaluation of different boronate affinity methods in their ability to retain ADP-ribose and pyridine moieties [75]. Using binding assays to compare the three different immobilized boronates of Affi-Gel 601, dihydroxyboryl-Sepharose, and dihydroxyboryl-Bio-Rex boronates, they showed that all three matrices were capable of binding free ADP-ribose with equal affinity, but only dihydroxyboryl-Bio-Rex was able to capture PAR efficiently. Additional binding experiments with MARylated histone H1 protein showed that this particular boronate matrix is unable to retain even 10% of the modified histone protein, indicating a potential bias towards PARylated over MARylated protein enrichment. As in earlier studies, for boronate to capture the cis-diol group in ADP-ribosylated proteins [73], a buffer of at least pH 8.5 is generally recommended [76]. Such basic pH conditions are necessary for the boronate to be hydroxylated and take on a tetrahedral anion geometry, which allows for the optimal interaction with the planar cis-diol group [76]. However, at these pH conditions, PAR and/or MAR may be chemically released from modified proteins by hydrolysis [74], likely reducing the amount of PARylated and/or MARylated proteins during the enrichment procedure. Another drawback of this approach is that boronate can bind to any biomolecules containing cis-diol groups, including RNA, carbohydrates, and glycoproteins. Therefore, boronate-enriched proteins cannot necessarily be considered ADP-ribosylated until the sites of ADP-ribosylation are definitively identified. The Yu group has recently successfully identified 340 proteins using boronate affinity chromatography combined with a hydroxylamine elution strategy, and defined 1,048 ADP-ribosylated sites at glutamate/aspartate residues in the human proteome ([77]; see Section 3.1 for details).
2.4 Use of NAD+ analogues
As ADP-ribosylation involves removal of the nicotinamide of NAD+ and transferring the remaining ADP-ribose moiety to the protein substrates, identification of ADP-ribosylated proteins has been facilitated by the use of NAD+ analogues, including biotinylated NAD+ [78-81], 1,N6etheno NAD+ (ε-NAD+) [82], and ‘clickable’ alkyne-NAD+ [83,84] (Figure 2). When coupled with specific PARPs, these NAD+ analogues allow for the determination of ADP-ribosylated substrates specific to individual enzymes [79,81,84].
2.4.1 Biotinylated NAD+
Since the ribose group within NAD+ is required for the ADP-ribosylation reaction with the nicotinamide moiety as the leaving group, the adenosine group is commonly modified to accommodate additional chemical groups. Zhang and Snyder have synthesized several NAD+ analogues biotinylated at the N6 or C8 positions and identified that the most reactive analogue requires 17 carbon spaces between the 6-position of adenine and biotin for efficient ADP-ribosylation, likely due to the bulkiness of the biotin group [85]; this analogue, designated 6-bio-17-NAD+, is currently commercially available. They further demonstrated that the biotinylated NAD+ can be used by diphtheria toxin to ADP-ribosylate the known substrate elongation factor 2; in combination with avidin affinity chromatography, they have identified glyceraldehyde-3-phophate dehydrogenase to be ADP-ribosylated upon nitric oxide stimulation in brain extract [85].
Recently, biotinylated NAD+ has been used in concert with protein microarrays as an approach to identify substrates of PARP2 and two mono(ADP-ribosyl)transferases ARTD10 (a.k.a. PARP10) and ARTD8 (PARP14) [79,81]. In these cases, the commercially available ProtoArrays® composed of a nitrocellulose membrane with about 8,000 proteins immobilized on the surface were used. Incubation of the ProtoArrays® with biotin NAD+ and individual ADP-ribosyltransferase allowed for the covalent labeling of PARylated (PARP2) and MARylated proteins (PARP10/PARP14), respectively, which were then detected by fluorophore-coupled streptavidin. Out of 8,000 proteins, the Rusconi group found 51 substrates of PARP2 while the Lüscher group found 46 unique substrates of PARP10 and 110 unique substrates of PARP14, with 32 substrates showing possible covalent modifications by both mono(ADP-ribosyl)transferases. The selection of less than 2% of proteins to be modified by each enzyme indicates the underlying substrate specificities of these ADP-ribosyltransferase family members. However, one of the major disadvantages of this method is the lack of consideration of the physiological context, both in terms of the level and localization of these enzymes and substrates being expressed in cells, and hence their endogenous stoichiometric ratios. Nevertheless, this proteome array-based approach allows a comprehensive survey of the possible substrates of specific ADP-ribosyltransferases or PARPs, and the relevance of these identified proteins can now be explored in cellular settings.
2.4.2 1,N6-etheno NAD+ (ε-NAD+)
The efficiency of PARylation has also been investigated with various NAD+ analogues, including those that have substitutions of the adenine moiety with either guanine, hypoxanthine or 1,N6-ethenoadenine [86]. Albeit with lower efficiency, these analogues can all serve as PARylation substrates. In particular in combination with a mouse monoclonal antibody 1G4 specific to ethenoadenosine [87], 1,N6-etheno NAD+ (ε-NAD+) has been used since the 1990s to detect PARP activity in permeabilized cells [88], as well as MARylation activity on the cell surface [89]. Recently, Guetg and colleagues (2012) have incubated ε-NAD+ with isolated nucleoli and identified that PARP1 and histones are ADP-ribosylated in this membrane-less organelle [82].
2.4.3 Clickable analogues of NAD+
Jiang and colleagues utilized click chemistry with an NAD+ analogue to label and identify 79 ADP-ribosylated substrates [83]. The NAD+ analogue possesses an alkyne group on the six position of the adenosine (6-alkyne-NAD+), allowing conjugation to a biotin tag via click chemistry to allow for isolation. To identify PARP1-specific substrates, the labeling reaction is performed by the addition of the NAD+ analogue to PARP1 knockdown cell lysates together with recombinant PARP1. Subsequent click chemistry allows the attachment of a Biotin-N3 affinity tag, and the labeled proteins are captured by streptavidin beads. The denaturing conditions of the labeling reaction ensure that only covalently modified proteins will be isolated, allowing for the distinction to be made between covalently modified ADP-ribosylated protein substrates and proteins that are non-covalently associated with ADP-ribose. Among the top hits were known substrates of PARP1 and ADP-ribosylated proteins previously identified in studies using the PAR-specific antibody 10H, validating this approach.
Recently the Cohen group further optimized the clickable NAD+ analogue and implemented a “bump-hole” strategy for engineering analogue-sensitive PARPs to identify PARP-specific substrates [84]. They engineered a range of substituents to create “bumps” on NAD+ at the C5-position of the nicotinamide moiety and tested whether they can fit into a “hole” made by converting a conserved lysine (K903) to alanine in PARP1. This particular lysine is present in all PARPs and forms van der Waals contacts with the C-5 position in the nicotinamide binding pocket of PARP1. From their screen, 5-ethyl-6-alkyne-NAD+ was identified to be selectively utilized by the engineered PARP1 but not by the wild-type up to a concentration of 250 μM, as demonstrated by the automodification reactions. A similar strategy was shown to be successful for PARP2 and PARP5b. However, one major drawback of the current approach is that the conserved lysine seems to be involved in PAR elongation and only MARylation was observed in all three analog-sensitive PARPs. Nonetheless, this enzyme-specific transfer of ADP-ribose onto target substrates enabled them to identify 42 and 301 substrates specific to PARP1 and PARP2, respectively, by LC-MS/MS.
2.4.4 Critical barrier – cell membrane
While use of NAD+ analogues provides the convenience of labeling and isolation of substrate proteins, this method of ADP-ribosylation analysis has the limitation of being restricted to in vitro or extracellular studies because NAD+ cannot cross membranes. One way to circumvent the barrier is to introduce NAD+ analogues into cells by using brief detergent permeabilization. This treatment has been successfully used for measuring PARP activities [88,90] and enriching for PARylated proteins for mass spectrometry analyses [80]. Given that these cells have been compromised, the investigation of their physiological relevance is warranted. Thus, the field will greatly benefit from an efficient method to deliver NAD+ analogues into cells.
2.5 Practical considerations for enriching PARylated proteins
Though PAR that is constitutively present in cells has a half-life of ~7.7 hours [91], PARylation can be rapidly induced and degraded by PARP and PARG respectively. During DNA damage, the increase in PARylation can be 10-500 fold but the half-life of the stress-induced polymer is very short (40 seconds to 6 minutes; reviewed in [92]). Therefore, it is critical to optimize the right time-points to profile PARylated proteins [44,45]. Given that PARG is particularly active in these stress conditions, the PARG inhibitor ADP-HPD should be added in the lysis buffer to preserve the elevated level of PARylated proteins [93]. Another approach to increase the amount of endogenous PARylated protein for identification is to genetically knock down PARG using siRNA or shRNA [55,77]. However, such sustained change in PAR turnover may disrupt cellular physiology and prevent the detection of the proper dynamic changes of PARylated proteins.
Besides preserving the PAR level due to PARG activity in cell lysate, it is equally important not to induce PARylation during experimental procedures. Jungmichel and colleagues recently identified that the DNA shearing during lysate preparation is sufficient to induce PARP activation [55]. Such non-physiological PARylation can be prevented by the addition of the PARP inhibitors 3-AB or PJ-34 in the lysis buffer [55].
3. SITE IDENTIFICATION
The burning question in the field of ADP-ribosylation proteomics is to identify the sites of modification. As discussed in Sections 2.1 and 2.2, this question becomes particularly important for enrichment methods that use 10H or ADP-ribose recognition modules because of their inabilities to discriminate ADP-ribosylated proteins from non-covalent binders. The identification of modified peptides with a characteristic mass shift will help define some of these enriched proteins to be ADP-ribosylated and determine the modification sites. For MARylated proteins, it is possible to detect a mass shift of 541.0611 Da for the addition of a single ADP-ribose group on a modified peptide as demonstrated by multiple studies on purified proteins or peptides [31,94-103]. Of note, multiple fragmentation methods including collision-induced dissociation, electron capture dissociation and electron transfer dissociation have been explored to identify the MARylated sites (recently reviewed in [39]). Reasoning that ADP-ribosylated peptides can be captured by phosphopeptides enrichment techniques [100], Matic and colleagues have been able to identify 79 MARylated proteins by re-analyzing existing phosphoproteomics data [101]. For PARylated proteins, it is however theoretically impossible to have a characteristic mass signature due to the heterogeneity of the polymer chain length (2-200 ADP-ribose units). Currently, three different methods have been proposed to reduce the chain length heterogeneity for identification by mass spectrometry using (1) hydroxylamine, (2) PARG/ARH3 and (3) phosphodiesterase (Figure 3).
3.1 Hydroxylamine-based approach
Hydroxylamine cleaves the ester bonds between the proximal ADP-ribose units of PAR and the carboxyl group of aspartate and glutamate residues, generating a characteristic 15.0109 Da mass signature on a modified site that can be readily detected by mass spectrometry [77]. This method has been used in conjugation with the boronate method to enrich for ADP-ribosylated proteins and identified 1,048 modified aspartate/glutamate sites from 340 proteins (see also Section 2.3). Using this approach, Zhang et al found that over 80% of sites are modified at glutamate, which have a marked tendency to reside on protein surfaces. They further defined a consensus motif surrounding the modified glutamate as PXE*, E*P, PXXE* and E*XXG. However, it should be noted that a sizeable fraction of endogenous ADP-ribosylated sites are not sensitive to hydroxylamine [33,104]. Therefore, other complementary approaches are needed to comprehensively profile the extent of the ADP-ribosylated proteome.
3.2 Use of PARG/ARH3
Given that it is technically feasible to identify MARylated sites [31,94-103], the Hottiger group used PARG to reduce the complexity of different PAR chain lengths on PARylated histone tail peptides [105]. The rationale behind this approach is that PARG can cleave the ribose-ribose bonds within the PAR chain, but not the covalent bonds between modified amino acids and the proximal ADP-ribose units [28], thereby essentially converting a PARylated site into a MARylated one for mass spectrometry identification. Similarly, since ARH3 can also cleave the ester bonds between ribose-ribose groups within the PAR chain [26,27], the Hottiger group reasoned that this degradation enzyme can also be used to identify PARylation sites [106]. Treating in vitro PARylated histone tail peptides with ARH3, they identified multiple modified lysine residues by mass spectrometry [106]. However, before furthering using ARH3 for site identification, the field will benefit from testing definitively on whether ARH3 will or will not remove the proximal ADP-ribose groups from PARylated proteins. One potential caveat of this approach is that both PARG and ARH3 generate free ADP-ribose upon cleaving the ribose-ribose bond from PAR chain. Given that free ADP-ribose has been shown to add onto proteins non-enzymatically at amino acid residues such as lysine and cysteine [107-109], it is currently not known whether the amount of free ADP-ribose generated during PARG/ARH3 cleavage will constitute a non-specific background in a proteome-wide experiment.
3.3 Phosphodiesterase-based approach
Another approach proposed to reduce PAR chain complexity is the use of a snake venom phosphodiesterase, which cleaves between the pyrophosphate bonds within the PAR chain [110]. This phosphodiesterase has traditionally been used to determine the length and structure of PAR [110,111] and was recently used for absolute quantification of PAR by mass spectrometry [112]. The complete cleavage of the pyrophosphate groups within the PAR chain leaves behind a unique mass tag of a phospho-ribose group attached to the modified amino acids (212.0086 Da). This method has been successfully demonstrated on purified peptides and proteins [9,31,39,65,113] and recently at the proteome level [65]. Daniels et al utilized immobilized metal affinity chromatography to enrich for phospho-ribose modified peptides and eluted them with a neutral phosphate buffer, rather than the standard ammonium hydroxide-based buffer to preserve the base-labile bonds between ADP-ribose group and peptide [65]. Currently, the coverage of the modified proteome is rather limited probably due to the inefficient cleavage by the phosphodiesterase within whole cell lysates. An additional consideration for utilizing this technique is that proteins have been shown to be endogenously modified with phospho-ribose groups [114], which were also identified in a recent re-analysis of a phosphoproteome data set [101]. Though the pathway to generate the protein modification with phospho-ribose and the function of such modification remain unknown, it is generally believed that the modification is derived from ADP-ribosylation [31,100]. Therefore, further investigation of this “derivative” protein modification is warranted.
4. CONCLUDING REMARKS
As we are still in the early days of analyzing the ADP-ribosylated proteome, it is not surprising that current efforts for substrate identification are far from reaching completion. Amongst the 1,548 unique proteins identified from three major studies using different approaches [45,55,77], only 190 proteins are overlapped in any two datasets and only 37 proteins overlap in all three studies. As illustrated by the Poirier group [45], ADP-ribosylated proteomes identified by Af1521 macrodomain, 10H antibody and PARG-DEAD approach were largely overlapped, but unique proteins were detected by each method. Currently, the extent and significance of the ADP-ribosylated proteome modified at different amino acids, including glutamate, aspartate, lysine and arginine, is still not known. Given that ADP-ribosylation can also be conjugated to cysteine, histidine and other amino acids [9,74], a non-biased proteomics approach to comprehensively identify all classes of amino acids is urgently needed. At the same time, pH conditions for boronate affinity chromatography and hydrolase activities for macrodomains should be optimized/altered so as not to reduce the inherent sensitivities of these enrichment methods (e.g. [115]). In addition, the field will greatly benefit from a more extensive exploration of the substrate specificities for macrodomain and other ADP-ribose binding domain containing proteins used for enrichment, as different substrate specificities may bias results.
As proteomics tools to enrich and identify ADP-ribosylated proteins are increasingly developed, several fundamental questions must be addressed: (1) What are the substrate specificities for PARPs, sirtuins and other mono(ADP-ribosyl)transferases? Are specific motifs recognized by these enzymes? (2) What are the roles of these ADP-ribosylated sites? (3) Are there specific functions for a single residue when it is MARylated or PARylated? A common theme emerging from existing large-scale ADP-ribosylated proteome studies is the enrichment of DNA-binding proteins and RNA-binding proteins [18,43,45,55,77]. Yet, data analyses have also revealed the possible novel roles of ADP-ribosylation in endosome trafficking and localization of nucleolar proteins and splicing factors [43,45,55,77]. Given that PARPs and sirtuins are active targets currently explored by pharmaceutical companies [13,116-118], systematic analyses on how these drugs modulate ADP-ribosylated proteomes will thus help delineate the observed clinical benefits and side effects.
Acknowledgements
We thank Dr. Shao-En Ong and Ms. Casey Daniels for providing input for the manuscript. This work was partly supported by a DOD Idea Award BC101881 (A.K.L.L), a pilot grant derived from the NIH grant U54GM103520 (A.K.L.L.) and an NCI training grant 5T32CA009110-35 (C.A.V).
Abbreviations
- ADP
ribose Adenosine Diphosphate-ribose
- AMP
Adenosine MonoPhosphate
- ARH3
ADP-ribosylhydrolase 3
- ε-NAD+
1,N6etheno-NAD+
- GFP
Green Fluorescent Protein
- IR
Ionizing radiation
- LC-MS/MS
Liquid Chromatography Mass Spectrometry/Mass Spectrometry
- MAR
Mono(ADP-ribose)
- MARylation
Mono(ADP-ribosyl)ation
- MALDI-TOF MS
Matrix assisted laser desorption/ionization- Time of flight mass spectrometry
- MNNG
N-methyl-N′-nitro-N-nitrosoguanidine
- NAD+
Nicotinamide Adenine Dinucleotide
- PAR
Poly(ADP-ribose)
- PARG
Poly(ADP-ribose) Glycohydrolase
- PARP
Poly(ADP-ribose) Polymerase
- PARylation
Poly(ADP-ribosyl)ation
- PBM
PAR Binding Motif
- PBZ
PAR Binding Zinc finger
References
- 1.Schreiber V, Dantzer F, Ame J-C, de Murcia G. de, Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol. 2006;7:517–528. doi: 10.1038/nrm1963. [DOI] [PubMed] [Google Scholar]
- 2.Hottiger MO, Hassa PO, Lüscher B, Schüler H, Koch-Nolte F. Toward a unified nomenclature for mammalian ADP-ribosyltransferases. Trends Biochem Sci. 2010 doi: 10.1016/j.tibs.2009.12.003. [DOI] [PubMed] [Google Scholar]
- 3.Di Girolamo M, Dani N, Stilla A, Corda D. Physiological relevance of the endogenous mono(ADP-ribosyl)ation of cellular proteins. FEBS J. 2005;272:4565–4575. doi: 10.1111/j.1742-4658.2005.04876.x. [DOI] [PubMed] [Google Scholar]
- 4.Koch-Nolte F, Kernstock S, Mueller-Dieckmann C, Weiss MS, Haag F. Mammalian ADP-ribosyltransferases and ADP-ribosylhydrolases. Front Biosci. 2008;13:6716–6729. doi: 10.2741/3184. [DOI] [PubMed] [Google Scholar]
- 5.Gibson BA, Kraus WL. New insights into the molecular and cellular functions of poly(ADP-ribose) and PARPs. Nat Rev Mol Cell Biol. 2012 doi: 10.1038/nrm3376. [DOI] [PubMed] [Google Scholar]
- 6.Krietsch J, Rouleau M, Pic E, Ethier C, et al. Reprogramming cellular events by poly(ADP-ribose)-binding proteins. Mol. Aspects Med. 2012 doi: 10.1016/j.mam.2012.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Karlberg T, Langelier M-F, Pascal JM, Schüler H. Structural biology of the writers, readers, and erasers in mono- and poly(ADP-ribose) mediated signaling. Mol. Aspects Med. 2013 doi: 10.1016/j.mam.2013.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bürkle A, Virág L. Poly(ADP-ribose): PARadigms and PARadoxes. Mol. Aspects Med. 2013;34:1046–1065. doi: 10.1016/j.mam.2012.12.010. [DOI] [PubMed] [Google Scholar]
- 9.Vyas S, Matic I, Uchima L, Rood JE, et al. Family-wide analysis of poly(ADP-ribose) polymerase activity. Nature Communications. 2014;5:4426. doi: 10.1038/ncomms5426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kickhoefer VA, Siva AC, Kedersha NL, Inman EM, et al. The 193-kD vault protein, VPARP, is a novel poly(ADP-ribose) polymerase. The Journal of Cell Biology. 1999;146:917–928. doi: 10.1083/jcb.146.5.917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Otto H, Reche PA, Bazan F, Dittmar K, et al. In silico characterization of the family of PARP-like poly(ADP-ribosyl)transferases (pARTs). BMC Genomics. 2005;6:139. doi: 10.1186/1471-2164-6-139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Perina D, Mikoč A, Ahel J, Cetković H, et al. Distribution of protein poly(ADP-ribosyl)ation systems across all domains of life. DNA Repair (Amst.) 2014 doi: 10.1016/j.dnarep.2014.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. PARP inhibition: PARP1 and beyond. Nat Rev Cancer. 2010;10:293–301. doi: 10.1038/nrc2812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Leung AKL, Vyas S, Rood JE, Bhutkar A, et al. Poly(ADP-ribose) regulates stress responses and microRNA activity in the cytoplasm. Mol Cell. 2011;42:489–499. doi: 10.1016/j.molcel.2011.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Briggs AG, Bent AF. Poly(ADP-ribosyl)ation in plants. Trends Plant Sci. 2011;16:372–380. doi: 10.1016/j.tplants.2011.03.008. [DOI] [PubMed] [Google Scholar]
- 16.Kim MY, Zhang T, Kraus WL. Poly(ADP-ribosyl)ation by PARP-1: “PAR-laying” NAD+ into a nuclear signal. Genes Dev. 2005;19:1951–1967. doi: 10.1101/gad.1331805. [DOI] [PubMed] [Google Scholar]
- 17.Jagtap P, Szabó C. Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors. Nature reviews Drug discovery. 2005;4:421–440. doi: 10.1038/nrd1718. [DOI] [PubMed] [Google Scholar]
- 18.Leung AKL. Poly(ADP-ribose): An organizer of cellular architecture. The Journal of Cell Biology. 2014;205:613–619. doi: 10.1083/jcb.201402114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fahrer J, Popp O, Malanga M, Beneke S, et al. High-affinity interaction of poly(ADP-ribose) and the human DEK oncoprotein depends upon chain length. Biochemistry. 2010;49:7119–7130. doi: 10.1021/bi1004365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Min W, Bruhn C, Grigaravicius P, Zhou Z-W, et al. Poly(ADP-ribose) binding to Chk1 at stalled replication forks is required for S-phase checkpoint activation. Nature Communications. 2013;4:2993. doi: 10.1038/ncomms3993. [DOI] [PubMed] [Google Scholar]
- 21.Ahel D, Horejsí Z, Wiechens N, Polo SE, et al. Poly(ADP-ribose)-dependent regulation of DNA repair by the chromatin remodeling enzyme ALC1. Science. 2009;325:1240–1243. doi: 10.1126/science.1177321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chang P, Jacobson MK, Mitchison TJ. Poly(ADP-ribose) is required for spindle assembly and structure. Nature. 2004;432:645–649. doi: 10.1038/nature03061. [DOI] [PubMed] [Google Scholar]
- 23.Hatakeyama K, Nemoto Y, Ueda K, Hayaishi O. Purification and characterization of poly(ADP-ribose) glycohydrolase. Different modes of action on large and small poly(ADP-ribose). J Biol Chem. 1986;261:14902–14911. [PubMed] [Google Scholar]
- 24.Meyer-Ficca ML, Meyer RG, Coyle DL, Jacobson EL, Jacobson MK. Human poly(ADP-ribose) glycohydrolase is expressed in alternative splice variants yielding isoforms that localize to different cell compartments. Exp Cell Res. 2004;297:521–532. doi: 10.1016/j.yexcr.2004.03.050. [DOI] [PubMed] [Google Scholar]
- 25.Niere M, Mashimo M, Agledal L, Dölle C, et al. ADP-ribosylhydrolase 3 (ARH3), not poly(ADP-ribose) glycohydrolase (PARG) isoforms, is responsible for degradation of mitochondrial matrix-associated poly(ADP-ribose). J Biol Chem. 2012;287:16088–16102. doi: 10.1074/jbc.M112.349183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Oka S, Kato J, Moss J. Identification and characterization of a mammalian 39-kDa poly(ADP-ribose) glycohydrolase. J Biol Chem. 2006;281:705–713. doi: 10.1074/jbc.M510290200. [DOI] [PubMed] [Google Scholar]
- 27.Mueller-Dieckmann C, Kernstock S, Lisurek M, Kries, von JP, et al. The structure of human ADP-ribosylhydrolase 3 (ARH3) provides insights into the reversibility of protein ADP-ribosylation. Proc Natl Acad Sci USA. 2006;103:15026–15031. doi: 10.1073/pnas.0606762103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Slade D, Dunstan MS, Barkauskaite E, Weston R, et al. The structure and catalytic mechanism of a poly(ADP-ribose) glycohydrolase. Nature. 2011 doi: 10.1038/nature10404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Rosenthal F, Feijs KLH, Frugier E, Bonalli M, et al. Macrodomain-containing proteins are new mono-ADP-ribosylhydrolases. Nat Struct Mol Biol. 2013 doi: 10.1038/nsmb.2521. [DOI] [PubMed] [Google Scholar]
- 30.Jankevicius G, Hassler M, Golia B, Rybin V, et al. A family of macrodomain proteins reverses cellular mono-ADP-ribosylation. Nat Struct Mol Biol. 2013 doi: 10.1038/nsmb.2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sharifi R, Morra R, Denise Appel C, Tallis M, et al. Deficiency of terminal ADP-ribose protein glycohydrolase TARG1/C6orf130 in neurodegenerative disease. EMBO J. 2013 doi: 10.1038/emboj.2013.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Bredehorst R, Wielckens K, Adamietz P, Steinhagen-Thiessen E, Hilz H. Mono(ADP-ribosyl)ation and poly(ADP-ribosyl)ation of proteins in developing liver and in hepatomas: relation of conjugate subfractions to metabolic competence and proliferation rates. Eur J Biochem. 1981;120:267–274. doi: 10.1111/j.1432-1033.1981.tb05699.x. [DOI] [PubMed] [Google Scholar]
- 33.Wielckens K, Bredehorst R, Adamietz P, Hilz H. Protein-bound polymeric and monomeric ADP-ribose residues in hepatic tissues. Comparative analyses using a new procedure for the quantification of poly(ADP-ribose). Eur J Biochem. 1981;117:69–74. doi: 10.1111/j.1432-1033.1981.tb06303.x. [DOI] [PubMed] [Google Scholar]
- 34.Jacobson MK, Payne DM, Alvarez-Gonzalez R, Juarez-Salinas H, et al. Determination of in vivo levels of polymeric and monomeric ADP-ribose by fluorescence methods. Methods in Enzymology. 1984;106:483–494. doi: 10.1016/0076-6879(84)06052-3. [DOI] [PubMed] [Google Scholar]
- 35.Finkel T, Deng C-X, Mostoslavsky R. Recent progress in the biology and physiology of sirtuins. Nature. 2009;460:587–591. doi: 10.1038/nature08197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Haigis MC, Mostoslavsky R, Haigis KM, Fahie K, et al. SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic beta cells. Cell. 2006;126:941–954. doi: 10.1016/j.cell.2006.06.057. [DOI] [PubMed] [Google Scholar]
- 37.Mao Z, Hine C, Tian X, Van Meter M, et al. SIRT6 promotes DNA repair under stress by activating PARP1. Science. 2011;332:1443–1446. doi: 10.1126/science.1202723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cantó C, Sauve AA, Bai P. Crosstalk between poly(ADP-ribose) polymerase and sirtuin enzymes. Mol. Aspects Med. 2013 doi: 10.1016/j.mam.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hengel SM, Goodlett DR. A Review of Tandem Mass Spectrometry Characterization of Adenosine Diphosphate-Ribosylated Peptides. Int J Mass Spectrom. 2012;312:114–121. doi: 10.1016/j.ijms.2011.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kawamitsu H, Hoshino H, Okada H, Miwa M, et al. Monoclonal antibodies to poly(adenosine diphosphate ribose) recognize different structures. Biochemistry. 1984;23:3771–3777. doi: 10.1021/bi00311a032. [DOI] [PubMed] [Google Scholar]
- 41.Fahrer J, Kranaster R, Altmeyer M, Marx A, Bürkle A. Quantitative analysis of the binding affinity of poly(ADP-ribose) to specific binding proteins as a function of chain length. Nucleic Acids Res. 2007;35:e143. doi: 10.1093/nar/gkm944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Petesch SJ, Lis JT. Activator-Induced Spread of Poly(ADP-Ribose) Polymerase Promotes Nucleosome Loss at Hsp70. Mol Cell. 2011 doi: 10.1016/j.molcel.2011.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gagné J-P, Isabelle M, Lo KS, Bourassa S, et al. Proteome-wide identification of poly(ADP-ribose) binding proteins and poly(ADP-ribose)-associated protein complexes. Nucleic Acids Res. 2008;36:6959–6976. doi: 10.1093/nar/gkn771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Isabelle M, Gagné J-P, Gallouzi I-E, Poirier GG. Quantitative proteomics and dynamic imaging reveal that G3BP-mediated stress granule assembly is poly(ADP-ribose)-dependent following exposure to MNNG-induced DNA alkylation. J Cell Sci. 2012;125:4555–4566. doi: 10.1242/jcs.106963. [DOI] [PubMed] [Google Scholar]
- 45.Gagné J-P, Pic E, Isabelle M, Krietsch J, et al. Quantitative proteomics profiling of the poly(ADP-ribose)-related response to genotoxic stress. Nucleic Acids Res. 2012;40:7788–7805. doi: 10.1093/nar/gks486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Realini CA, Althaus FR. Histone shuttling by poly(ADP-ribosylation). J Biol Chem. 1992;267:18858–18865. [PubMed] [Google Scholar]
- 47.Haince J-F, Poirier GG, Kirkland JB. Nonisotopic methods for determination of poly(ADP-ribose) levels and detection of poly(ADP-ribose) polymerase. Curr Protoc Cell Biol. 2004 doi: 10.1002/0471143030.cb1807s21. Chapter 18, Unit18.7. [DOI] [PubMed] [Google Scholar]
- 48.Schwab CJ, Colville MJ, Fullerton AT, McMahon KK. Evidence of endogenous mono-ADP-ribosylation of cardiac proteins via anti-ADP-ribosylarginine immunoreactivity. Proc. Soc. Exp. Biol. Med. 2000;223:389–396. doi: 10.1046/j.1525-1373.2000.22355.x. [DOI] [PubMed] [Google Scholar]
- 49.Meyer T, Hilz H. Production of anti-(ADP-ribose) antibodies with the aid of a dinucleotide-pyrophosphatase-resistant hapten and their application for the detection of mono(ADP-ribosyl)ated polypeptides. Eur J Biochem. 1986;155:157–165. doi: 10.1111/j.1432-1033.1986.tb09471.x. [DOI] [PubMed] [Google Scholar]
- 50.Osago H, Terashima M, Hara N, Yamada K, Tsuchiya M. A new detection method for arginine-specific ADP-ribosylation of protein -- a combinational use of anti-ADP-ribosylarginine antibody and ADP-ribosylarginine hydrolase. J. Biochem. Biophys. Methods. 2008;70:1014–1019. doi: 10.1016/j.jprot.2007.11.008. [DOI] [PubMed] [Google Scholar]
- 51.Siegmund KD, Klink F. Production of an antiserum specific to the ADP-ribosylated form of elongation factor 2 from archaebacteria and eukaryotes. FEBS Lett. 1992;312:139–142. doi: 10.1016/0014-5793(92)80921-3. [DOI] [PubMed] [Google Scholar]
- 52.Eide B, Gierschik P, Spiegel A. Immunochemical detection of guanine nucleotide binding proteins mono-ADP-ribosylated by bacterial toxins. Biochemistry. 1986;25:6711–6715. doi: 10.1021/bi00369a058. [DOI] [PubMed] [Google Scholar]
- 53.Kalisch T, Ame J-C, Dantzer F, Schreiber V. New readers and interpretations of poly(ADP-ribosyl)ation. Trends Biochem Sci. 2012;37:381–390. doi: 10.1016/j.tibs.2012.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Dani N, Stilla A, Marchegiani A, Tamburro A, et al. Combining affinity purification by ADP-ribose-binding macro domains with mass spectrometry to define the mammalian ADP-ribosyl proteome. Proc Natl Acad Sci USA. 2009;106:4243–4248. doi: 10.1073/pnas.0900066106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jungmichel S, Rosenthal F, Altmeyer M, Lukas J, et al. Proteome-wide Identification of Poly(ADP-Ribosyl)ation Targets in Different Genotoxic Stress Responses. Mol Cell. 2013 doi: 10.1016/j.molcel.2013.08.026. [DOI] [PubMed] [Google Scholar]
- 56.Pehrson JR, Fried VA. MacroH2A, a core histone containing a large nonhistone region. Science. 1992;257:1398–1400. doi: 10.1126/science.1529340. [DOI] [PubMed] [Google Scholar]
- 57.Feijs KLH, Forst AH, Verheugd P, Lüscher B. Macrodomain-containing proteins: regulating new intracellular functions of mono(ADP-ribosyl)ation. Nat Rev Mol Cell Biol. 2013;14:445–453. doi: 10.1038/nrm3601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chen D, Vollmar M, Rossi MN, Phillips C, et al. Identification of macrodomain proteins as novel O-acetyl-ADP-ribose deacetylases. J Biol Chem. 2011;286:13261–13271. doi: 10.1074/jbc.M110.206771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Allen MD, Buckle AM, Cordell SC, Löwe J, Bycroft M. The crystal structure of AF1521 a protein from Archaeoglobus fulgidus with homology to the non-histone domain of macroH2A. J Mol Biol. 2003;330:503–511. doi: 10.1016/s0022-2836(03)00473-x. [DOI] [PubMed] [Google Scholar]
- 60.Kustatscher G, Hothorn M, Pugieux C, Scheffzek K, Ladurner AG. Splicing regulates NAD metabolite binding to histone macroH2A. Nat Struct Mol Biol. 2005;12:624–625. doi: 10.1038/nsmb956. [DOI] [PubMed] [Google Scholar]
- 61.Timinszky G, Till S, Hassa PO, Hothorn M, et al. A macrodomain-containing histone rearranges chromatin upon sensing PARP1 activation. Nat Struct Mol Biol. 2009;16:923–929. doi: 10.1038/nsmb.1664. [DOI] [PubMed] [Google Scholar]
- 62.Forst AH, Karlberg T, Herzog N, Thorsell A-G, et al. Recognition of mono-ADP-ribosylated ARTD10 substrates by ARTD8 macrodomains. Structure. 2013;21:462–475. doi: 10.1016/j.str.2012.12.019. [DOI] [PubMed] [Google Scholar]
- 63.Karras GI, Kustatscher G, Buhecha HR, Allen MD, et al. The macro domain is an ADP-ribose binding module. EMBO J. 2005;24:1911–1920. doi: 10.1038/sj.emboj.7600664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ong S-E, Blagoev B, Kratchmarova I, Kristensen DB, et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol Cell Proteomics. 2002;1:376–386. doi: 10.1074/mcp.m200025-mcp200. [DOI] [PubMed] [Google Scholar]
- 65.Daniels CM, Ong S-E, Leung AKL. Phosphoproteomic Approach to Characterize Protein Mono- and Poly(ADP-ribosyl)ation Sites from Cells. J Proteome Res. 2014 doi: 10.1021/pr401032q. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kim I-K, Kiefer JR, Ho CMW, Stegeman RA, et al. Structure of mammalian poly(ADP-ribose) glycohydrolase reveals a flexible tyrosine clasp as a substrate-binding element. Nat Struct Mol Biol. 2012;19:653–656. doi: 10.1038/nsmb.2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wang Z, Gagné J-P, Poirier GG, Xu W. Crystallographic and biochemical analysis of the mouse poly(ADP-ribose) glycohydrolase. PLoS ONE. 2014;9:e86010. doi: 10.1371/journal.pone.0086010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Moyle PM, Muir TW. Method for the synthesis of mono-ADP-ribose conjugated peptides. J Am Chem Soc. 2010;132:15878–15880. doi: 10.1021/ja1064312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kang HC, Lee Y-I, Shin J-H, Andrabi SA, et al. Iduna is a poly(ADP-ribose) (PAR)-dependent E3 ubiquitin ligase that regulates DNA damage. Proc Natl Acad Sci USA. 2011;108:14103–14108. doi: 10.1073/pnas.1108799108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Zhang Y, Liu S, Mickanin C, Feng Y, et al. RNF146 is a poly(ADP-ribose)-directed E3 ligase that regulates axin degradation and Wnt signalling. Nat Cell Biol. 2011;13:623–629. doi: 10.1038/ncb2222. [DOI] [PubMed] [Google Scholar]
- 71.Callow MG, Tran H, Phu L, Lau T, et al. Ubiquitin ligase RNF146 regulates tankyrase and Axin to promote Wnt signaling. PLoS ONE. 2011;6:e22595. doi: 10.1371/journal.pone.0022595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wang Z, Michaud GA, Cheng Z, Zhang Y, et al. Recognition of the iso-ADP-ribose moiety in poly(ADP-ribose) by WWE domains suggests a general mechanism for poly(ADP-ribosyl)ation-dependent ubiquitination. Genes Dev. 2012 doi: 10.1101/gad.182618.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Okayama H, Ueda K, Hayaishi O. Purification of ADP-ribosylated nuclear proteins by covalent chromatography on dihydroxyboryl polyacrylamide beads and their characterization. Proc Natl Acad Sci USA. 1978;75:1111–1115. doi: 10.1073/pnas.75.3.1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cervantes-Laurean D, Jacobson EL, Jacobson MK. Preparation of low molecular weight model conjugates for ADP-ribose linkages to protein. Methods in Enzymology. 1997;280:275–287. doi: 10.1016/s0076-6879(97)80119-x. [DOI] [PubMed] [Google Scholar]
- 75.Alvarez-Gonzalez R, Juarez-Salinas H, Jacobson EL, Jacobson MK. Evaluation of immobilized boronates for studies of adenine and pyridine nucleotide metabolism. Anal Biochem. 1983;135:69–77. doi: 10.1016/0003-2697(83)90732-7. [DOI] [PubMed] [Google Scholar]
- 76.Liu XC, Scouten WH. Boronate affinity chromatography. Methods Mol Biol. 2000;147:119–128. doi: 10.1007/978-1-60327-261-2_12. [DOI] [PubMed] [Google Scholar]
- 77.Zhang Y, Wang J, Ding M, Yu Y. Site-specific characterization of the Asp- and Glu-ADP-ribosylated proteome. Nat Methods. 2013 doi: 10.1038/nmeth.2603. [DOI] [PubMed] [Google Scholar]
- 78.Zhang J. Use of biotinylated NAD to label and purify ADP-ribosylated proteins. Methods in Enzymology. 1997;280:255–265. doi: 10.1016/s0076-6879(97)80117-6. [DOI] [PubMed] [Google Scholar]
- 79.Feijs KL, Kleine H, Braczynski A, Forst AH, et al. ARTD10 substrate identification on protein microarrays: regulation of GSK3β by mono-ADP-ribosylation. Cell Commun. Signal. 2013;11:5. doi: 10.1186/1478-811X-11-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Farrar D, Rai S, Chernukhin I, Jagodic M, et al. Mutational analysis of the poly(ADP-ribosyl)ation sites of the transcription factor CTCF provides an insight into the mechanism of its regulation by poly(ADP-ribosyl)ation. Mol Cell Biol. 2010;30:1199–1216. doi: 10.1128/MCB.00827-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Troiani S, Lupi R, Perego R, Depaolini SR, et al. Identification of candidate substrates for poly(ADP-ribose) polymerase-2 (PARP2) in the absence of DNA damage using high-density protein microarrays. FEBS J. 2011;278:3676–3687. doi: 10.1111/j.1742-4658.2011.08286.x. [DOI] [PubMed] [Google Scholar]
- 82.Guetg C, Scheifele F, Rosenthal F, Hottiger MO, Santoro R. Inheritance of silent rDNA chromatin is mediated by PARP1 via noncoding RNA. Mol Cell. 2012;45:790–800. doi: 10.1016/j.molcel.2012.01.024. [DOI] [PubMed] [Google Scholar]
- 83.Jiang H, Kim JH, Frizzell KM, Kraus WL, Lin H. Clickable NAD analogues for labeling substrate proteins of poly(ADP-ribose) polymerases. J Am Chem Soc. 2010;132:9363–9372. doi: 10.1021/ja101588r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Carter-O'Connell IO, Jin H, Morgan RK, David LL, Cohen MS. Engineering the substrate specificity of ADP-ribosyltransferases for identifying direct protein targets. J Am Chem Soc. 2014 doi: 10.1021/ja412897a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhang J, Snyder SH. Purification of a nitric oxide-stimulated ADP-ribosylated protein using biotinylated beta-nicotinamide adenine dinucleotide. Biochemistry. 1993;32:2228–2233. doi: 10.1021/bi00060a014. [DOI] [PubMed] [Google Scholar]
- 86.Oei SL, Griesenbeck J, Buchlow G, Jorcke D, et al. NAD+ analogs substituted in the purine base as substrates for poly(ADP-ribosyl) transferase. FEBS Lett. 1996;397:17–21. doi: 10.1016/s0014-5793(96)01137-4. [DOI] [PubMed] [Google Scholar]
- 87.Young TL, Santella RM. Development of techniques to monitor for exposure to vinyl chloride: monoclonal antibodies to ethenoadenosine and ethenocytidine. Carcinogenesis. 1988;9:589–592. doi: 10.1093/carcin/9.4.589. [DOI] [PubMed] [Google Scholar]
- 88.Davis RE, Mysore V, Browning JC, Hsieh JC, et al. In situ staining for poly(ADP-ribose) polymerase activity using an NAD analogue. J Histochem Cytochem. 1998;46:1279–1289. doi: 10.1177/002215549804601108. [DOI] [PubMed] [Google Scholar]
- 89.Krebs C, Koestner W, Nissen M, Welge V, et al. Flow cytometric and immunoblot assays for cell surface ADP-ribosylation using a monoclonal antibody specific for ethenoadenosine. Anal Biochem. 2003;314:108–115. doi: 10.1016/s0003-2697(02)00640-1. [DOI] [PubMed] [Google Scholar]
- 90.Bakondi E, Bai P, Szabó E,E, Hunyadi J, et al. Detection of poly(ADP-ribose) polymerase activation in oxidatively stressed cells and tissues using biotinylated NAD substrate. J Histochem Cytochem. 2002;50:91–98. doi: 10.1177/002215540205000110. [DOI] [PubMed] [Google Scholar]
- 91.Alvarez-Gonzalez R, Althaus FR. Poly(ADP-ribose) catabolism in mammalian cells exposed to DNA-damaging agents. Mutat Res. 1989;218:67–74. doi: 10.1016/0921-8777(89)90012-8. [DOI] [PubMed] [Google Scholar]
- 92.Hassa PO, Hottiger MO. The diverse biological roles of mammalian PARPS, a small but powerful family of poly-ADP-ribose polymerases. Front Biosci. 2008;13:3046–3082. doi: 10.2741/2909. [DOI] [PubMed] [Google Scholar]
- 93.Slama JT, Aboul-Ela N, Jacobson MK. Mechanism of inhibition of poly(ADP-ribose) glycohydrolase by adenosine diphosphate (hydroxymethyl)pyrrolidinediol. J Med Chem. 1995;38:4332–4336. doi: 10.1021/jm00021a023. [DOI] [PubMed] [Google Scholar]
- 94.Hengel SM, Icenogle L, Collins C, Goodlett DR. Sequence assignment of ADP-ribosylated peptides is facilitated as peptide length increases. Rapid Commun. Mass Spectrom. 2010;24:2312–2316. doi: 10.1002/rcm.4600. [DOI] [PubMed] [Google Scholar]
- 95.Zee BM, Garcia BA. Electron transfer dissociation facilitates sequencing of adenosine diphosphate-ribosylated peptides. Anal. Chem. 2010;82:28–31. doi: 10.1021/ac902134y. [DOI] [PubMed] [Google Scholar]
- 96.Fedorova M, Frolov A, Hoffmann R. Fragmentation behavior of Amadori-peptides obtained by non-enzymatic glycosylation of lysine residues with ADP-ribose in tandem mass spectrometry. J Mass Spectrom. 2010;45:664–669. doi: 10.1002/jms.1758. [DOI] [PubMed] [Google Scholar]
- 97.Tao Z, Gao P, Liu H-W. Identification of the ADP-ribosylation sites in the PARP-1 automodification domain: analysis and implications. J Am Chem Soc. 2009;131:14258–14260. doi: 10.1021/ja906135d. [DOI] [PubMed] [Google Scholar]
- 98.Hengel SM, Shaffer SA, Nunn BL, Goodlett DR. Tandem mass spectrometry investigation of ADP-ribosylated kemptide. J. Am. Soc. Mass Spectrom. 2009;20:477–483. doi: 10.1016/j.jasms.2008.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Margarit SM, Davidson W, Frego L, Stebbins CE. A steric antagonism of actin polymerization by a salmonella virulence protein. Structure. 2006;14:1219–1229. doi: 10.1016/j.str.2006.05.022. [DOI] [PubMed] [Google Scholar]
- 100.Laing S, Koch-Nolte F, Haag F, Buck F. Strategies for the identification of arginine ADP-ribosylation sites. J Proteomics. 2011;75:169–176. doi: 10.1016/j.jprot.2011.07.003. [DOI] [PubMed] [Google Scholar]
- 101.Matic I, Ahel I, Hay RT. Reanalysis of phosphoproteomics data uncovers ADP-ribosylation sites. Nat Methods. 2012;9:771–772. doi: 10.1038/nmeth.2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Oetjen J, Rexroth S, Reinhold-Hurek B. Mass spectrometric characterization of the covalent modification of the nitrogenase Fe-protein in Azoarcus sp. BH72. FEBS J. 2009;276:3618–3627. doi: 10.1111/j.1742-4658.2009.07081.x. [DOI] [PubMed] [Google Scholar]
- 103.Jiang H, Sherwood R, Zhang S, Zhu X, et al. Identification of ADP-ribosylation sites of CD38 mutants by precursor ion scanning mass spectrometry. Anal Biochem. 2013;433:218–226. doi: 10.1016/j.ab.2012.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Adamietz P, Hilz H. Poly(adenosine diphosphate ribose) is covalently linked to nuclear proteins by two types of bonds. Hoppe-Seyler's Z Physiol Chem. 1976;357:527–534. doi: 10.1515/bchm2.1976.357.1.527. [DOI] [PubMed] [Google Scholar]
- 105.Rosenthal F, Messner S, Roschitzki B, Gehrig P, et al. Identification of distinct amino acids as ADP-ribose acceptor sites by mass spectrometry. Methods Mol Biol. 2011;780:57–66. doi: 10.1007/978-1-61779-270-0_4. [DOI] [PubMed] [Google Scholar]
- 106.Messner S, Altmeyer M, Zhao H, Pozivil A, et al. PARP1 ADP-ribosylates lysine residues of the core histone tails. Nucleic Acids Res. 2010;38:6350–6362. doi: 10.1093/nar/gkq463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kharadia SV, Graves DJ. Relationship of phosphorylation and ADP-ribosylation using a synthetic peptide as a model substrate. J Biol Chem. 1987;262:17379–17383. [PubMed] [Google Scholar]
- 108.McDonald LJ, Moss J. Enzymatic and nonenzymatic ADP-ribosylation of cysteine. Mol Cell Biochem. 1994;138:221–226. doi: 10.1007/BF00928465. [DOI] [PubMed] [Google Scholar]
- 109.Kun E, Chang AC, Sharma ML, Ferro AM, Nitecki D. Covalent modification of proteins by metabolites of NAD+. Proc Natl Acad Sci USA. 1976;73:3131–3135. doi: 10.1073/pnas.73.9.3131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Matsubara H, Hasegawa S, Fujimura S, Shima T, Sugimura T. Studies on poly (adenosine diphosphate ribose). V. Mechanism of hydrolysis of poly (adenosine diphosphate ribose) by snake venom phosphodiesterase. J Biol Chem. 1970;245:3606–3611. [PubMed] [Google Scholar]
- 111.Miwa M, Saikawa N, Yamaizumi Z, Nishimura S, Sugimura T. Structure of poly(adenosine diphosphate ribose): identification of 2′-[1″-ribosyl-2″-(or 3″-)(1‴-ribosyl)]adenosine-5′,5″,5′“−”tris(phosphate) as a branch linkage. Proc Natl Acad Sci USA. 1979;76:595–599. doi: 10.1073/pnas.76.2.595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Martello R, Mangerich A, Sass S, Dedon PC, Bürkle A. Quantification of cellular poly(ADP-ribosyl)ation by stable isotope dilution mass spectrometry reveals tissue- and drug-dependent stress response dynamics. ACS Chem. Biol. 2013 doi: 10.1021/cb400170b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Chapman JD, Gagné J-P, Poirier GG, Goodlett DR. Mapping PARP-1 auto-ADP-ribosylation sites by liquid chromatography-tandem mass spectrometry. J Proteome Res. 2013 doi: 10.1021/pr301219h. [DOI] [PubMed] [Google Scholar]
- 114.Williams JC, Chambers JP, Liehr JG. Glutamyl ribose 5-phosphate storage disease. A hereditary defect in the degradation of poly(ADP-ribosylated) proteins. J Biol Chem. 1984;259:1037–1042. [PubMed] [Google Scholar]
- 115.Liu Y, Ren L, Liu Z. A unique boronic acid functionalized monolithic capillary for specific capture, separation and immobilization of cis-diol biomolecules. Chem. Commun. (Camb.) 2011;47:5067–5069. doi: 10.1039/c0cc05675h. [DOI] [PubMed] [Google Scholar]
- 116.Alcaín FJ, Villalba JM. Sirtuin inhibitors. Expert Opin Ther Pat. 2009;19:283–294. doi: 10.1517/13543770902755111. [DOI] [PubMed] [Google Scholar]
- 117.Beaudeux J-L, Nivet-Antoine V, Giral P. Resveratrol: a relevant pharmacological approach for the treatment of metabolic syndrome? Curr Opin Clin Nutr Metab Care. 2010;13:729–736. doi: 10.1097/MCO.0b013e32833ef291. [DOI] [PubMed] [Google Scholar]
- 118.Garber K. PARP inhibitors bounce back. Nature reviews Drug discovery. 2013;12:725–727. doi: 10.1038/nrd4147. [DOI] [PubMed] [Google Scholar]