Abstract
PARP family members can be found spread across all domains and continue to be essential molecules from lower to higher eukaryotes. Poly (ADP‐ribose) polymerase 1 (PARP‐1), newly termed ADP‐ribosyltransferase D‐type 1 (ARTD1), is a ubiquitously expressed ADP‐ribosyltransferase (ART) enzyme involved in key cellular processes such as DNA repair and cell death. This review assesses current developments in PARP‐1 biology and activation signals for PARP‐1, other than conventional DNA damage activation. Moreover, many essential functions of PARP‐1 still remain elusive. PARP‐1 is found to be involved in a myriad of cellular events via conservation of genomic integrity, chromatin dynamics and transcriptional regulation. This article briefly focuses on its other equally important overlooked functions during growth, metabolic regulation, spermatogenesis, embryogenesis, epigenetics and differentiation. Understanding the role of PARP‐1, its multidimensional regulatory mechanisms in the cell and its dysregulation resulting in diseased states, will help in harnessing its true therapeutic potential.
1. Introduction
Poly (ADP‐ribose) polymerase (PARP) enzymes are a family of proteins involved in a number of cellular processes including gene regulation, chromatin remodelling, DNA repair and apoptosis.1 These enzymes are present in all eukaryotes except yeast.2 PARPs can either transfer a single unit of (ADP‐ribose) or more than one (ADP‐ribose) moieties from NAD+onto substrates yielding poly (ADP‐ribose) (PAR) chains, which can be of varying length and branch content. ADP‐ribosyltransferase D‐type 1 (ARTD‐1 or PARP‐1) falls in the latter category.1, 3 The PAR polymers are rapidly degraded by poly (ADP‐ribose) glycohydrolase (PARG)4 possessing both endoglycosidic and exoglycosidic activities,5 and PAR hydrolase (ARH3), which also shares catalytic domain similarity with PARG.6 However, ARH3 does not hydrolyse ADP‐ribose‐arginine, ‐cysteine, ‐diphthamide or ‐asparagine bonds.6 Another set of enzymes known as macro domain‐containing proteins and NUDIX hyrolases have also been reported to be involved in PAR degradation.7, 8 There are 17 different homologues of PARP that have a conserved catalytic domain with various domains like zinc finger, BRCT, SAM, SAP, ankyrin and macro domain.9 Though PARP‐1 has been demonstrated as a key player in DNA repair and cell death, many of its equally vital cellular functions have been overlooked. In this review, we discuss the distribution of PARP homologues across all organisms and the role of PARP‐1 in various cellular functions like transcription, spermatogenesis, epigenetics and the most novel in differentiation and multicellularity.
2. The PARP family
Based on new proposed nomenclature by Hottiger et al., 10 the human PARP (hPARP) family is classified into three groups depending on their motifs and functions: (1) PARP 1‐5: have a conserved glutamate residue (Glu988); (2) PARP 6‐8, 10‐12 and 14‐16: are putative mono‐(ADP‐ribose) polymerases and (3) PARP 9 and 13 which do not have PARP signature motif that binds NAD+nor do they have Glu988 implying that they are inactive.10 PARP superfamily can be subdivided into six clades which are shown in Table 1.
Table 1.
Distribution of PARP. PARP has been divided into six clades depending on the domains present22, 29 and 30
| Clade | Clade sub group | Class | Key features |
|---|---|---|---|
| Clade 1 | Clade 1A | Amoebozoa (Dictyostelium) Opisthokonta (Fungi) Chromalveolates | Ankyrin repeats, WGR PRD, PARP catalytic domains. |
| Clade 1B | Opisthokonta (animals and Choanoflagellata) and the Excavata (the Heterolobosea member Naegleria) | three N‐terminal zinc fingers that contribute to DNA binding, a BRCT domain and a PADR1 domain in addition to WGR, PRD and the catalytic domain | |
| Clade 1C | Oomyocete Phytophtora species (within the Excavata) and one basal animal. | WGR, PRD and PARP catalytic domains and mostly do not contain other functional domains. | |
| Clade1D | Opisthokonta, the animals Xenopus laevis (Q566G1) and Schistosoma japonicum (Q5DAZ0) and the fungus Batrachochytrium dendrobatidis and Plantae (land plants) as well as ciliate members of the Chromalveolates. | WGR, PRD and PARP catalytic domains and mostly do not contain other functional domains. | |
| Clade 1E | most of the fungal members of Clade 1 | BRCT domains N‐terminal to WGR, PRD and PARP catalytic domains. | |
| Clade 1F | the Excavata | — | |
| Clade 1G | Opisthokonta (both animals and the Choanoflagellate Monosiga brevicollis) | only WGR, PRD and PARP catalytic domains | |
| Clade 1H | Two Caenorhabditis elegans (C. elegans) proteins | PADR1, WGR, PRD and PARP | |
| Clade2 | Clade 2A | — | an N‐terminal WWE domain, the PARP signature and a C‐terminal extension |
| Clade 2B | — | only the PARP signature and the C‐terminal extension | |
| Clade 3 | Clade 3A | — | RRM RNA‐binding domain, a glycine‐rich region (GRD), and a UIM domain |
| Clade 3B | Trichoplax adhaerens | Macro domain N‐terminal to their C‐terminal catalytic domain | |
| Clade 3C | — | Macro domain N‐terminal to their C‐terminal catalytic domain | |
| Clade 3D | two Dictyostelium discoideum and four Tetrahymena thermophila proteins | — | |
| Clade 3E | — | one to two WWE domains, alone or in combination with zinc fingers (either CCCH or CCCH types) in front of their PARP catalytic domains | |
| Clade3F | — | PARP9 | |
| Clade 4 | Clade 4 | — | 15–18 ankyrin repeats followed by a sterile alpha motif (SAM) and the PARP catalytic domain |
| Clade 5 | Clade 5A | Opishthokonts (animals) | the PARP signature is found in the middle of the protein, rather than at the C terminus |
| Clade 5B | Amoebozoa | — | |
| Clade 6 | Clade 6A | Opisthokonts (animals and fungi), Excavates (Parabasalids and Heterolobosa), and Plantae (chlorophyta and bryophytes) | N termini with no known functional domains and C‐terminal extensions beyond the PARP catalytic domain of varying lengths |
| Clade 6B | PfamB_2311 domains as well as the PARP catalytic domain | ||
| Clade 6C | PfamB_2311 domain and a PARP catalytic domain | ||
| Clade 6D | Deuterostomes with the exception of the mollusc Lottia gigantean | PfamB_2311 domain and the PARP catalytic domain | |
| Clade 6E | seven proteins encoded by Trichomonas vaginalis | PfamB_2311 domain and the PARP catalytic domain |
Out of 17 members, PARP‐1 (113 kDa) was the first characterized and extensively studied enzyme recognized to play an essential role in DNA repair.11 PARP‐1 and PARP‐2 share ~69% homology in the catalytic domain and they are documented as vital proteins in DNA repair system,2 while PARP‐3 is reported to be a mono‐ADP‐ribosylating enzyme by Loseva and group.12 PARP‐2 and PARP‐3 were considered as a subgroup of PARP‐1 as they all carry out synthesis of branched polymers.13 PARP‐4 also known as Vault PARP, is a ribonucleoprotein complex having PARylation activity and it is thought to be involved in multidrug resistance of tumour and intracellular transport.14 Tankyrase‐1 (TRF‐1‐interacting ankyrin‐related ADP‐ribose polymerase‐1), also known as PARP5a, is identified to enhance telomere elongation by telomerase.15 Other PARP homologues show structural and functional differences. Tankyrase‐2 lacks N‐terminal HPS (His‐Pro‐Ser) domain, but it may share some overlapping functions with tankyrase‐1.16 Other PARP family members like tiPARP, PARP‐12 and PARP‐13 share PARP catalytic, WWE and CX8CX5CX3‐like zinc finger domains.2 PARP‐13 has been reported to be an important regulator of cellular mRNA via regulation of miRNA activity.17 The next subgroup which includes PARP‐9/BAL1, PARP‐14/BAL2/CoaSt6 and PARP‐15/BAL3 are macro‐PARPs, characterized by macro domains positioned before the PARP domain. This domain is found to be involved in transcriptional repression and X‐chromosome inactivation, suggesting it as a transcription factor.18 The RNA recognition motif (RRM) and the Gly‐rich domain of PARP‐10 are known to help in binding of RNA with proto‐oncoprotein c‐Myc.2 Other PARP family members such as PARP‐6, PARP‐8, PARP‐11 and PARP‐16 have been identified but their functions are still elusive, though PARP‐8 and 16 have been recently shown to be involved in assembly or maintenance of membranous organelles.19
3. Distribution of PARP across life
3.1. PARP in lower life forms
3.1.1. PARP in bacteria
Numerous PARP‐like proteins are detected in several bacterial genomes.20, 21 Till now, around 28 PARP homologues have been suggested across 27 bacterial species.22 However, only a few bacteria possess the entire machinery required for PARP metabolism. Some also show the conserved histidine‐tyrosine‐glutamate (H‐Y‐E) catalytic triad which is essential for its activity.21 PARP from Herpetosiphon aurantiacus has been reported to have conserved catalytic triad having the same characteristics as human PARP‐1 enzyme.21
3.1.2. PARP in archaea
Archaea do show the presence of PARP homologues. PARP‐like thermozymes have been identified from Sulfolobus solfataricus. This PARP‐like protein shows oligo (ADP‐ribosyl) transferase activity and DNA‐binding activity.23
3.1.3. PARP in viruses
PARP‐like proteins have also been identified in a few double‐stranded DNA viruses 24 such as Aeromonas phage—Aeh1, Anticarsia gemmatalis nucleopolyhedro virus, invertebrate iridescent virus 6 and cellulophagaphage phi4:1. All these viral PARPs have been found to possess the conserved catalytic triad H‐Y‐E with an exception of one which has an aspartate instead of glutamate suggesting that these PARPs are active ADP‐ribosyl transferases. Some viruses such as Herpes simplex virus and Epstein‐Barr virus have also been reported to use PAR metabolism for their replication.25, 26
3.2. PARP in higher eukaryotes
PARPs are found in a divergent group of eukaryotes.9, 10 PARP expression has been identified in nearly all eukaryotic cells ranging from plants to vertebrates.27 PARP‐1 was long assumed to be the single enzyme with PARylation function until two PARP isoforms were discovered in plants.28 Citarelli et al.29 investigated at least two more PARP proteins in the last common extant ancestor of eukaryotes.
In conclusion, it is clear that the complexity of PARP proteins is augmented with the evolutionary level of the species. Vyas et al.19 evidently illustrated that this domain complexity confers the diversity in functions to the PARP family.
PARP‐1 is best studied out of this 17‐member family of hPARPs. PARP has been implicated in development and cell differentiation from lower life forms to higher eukaryotes.30 However, it is involved in a plethora of functions and many of its functions in spermatogenesis, epigenetics and differentiation remain unclear. Thus, understanding PARP‐1 and its role in the above processes is the focus of this review.
4. PARP‐1: Structure, activation signals and its diverse cellular roles
4.1. Gene organization of PARP‐1 and its modifications
PARP‐1 (EC 2.4.2.30) is a prominent member of the PARP family. It is a nuclear enzyme with approximately 106 molecules per cell 31 and accounts for 80%–90% of total cellular PARylation. Gene structure of PARP‐1 mainly consists of DNA binding, an auto modification and a catalytic domain (Fig. 1). (1) The N‐terminal DNA‐binding domain has three zinc fingers and a nuclear localization sequence (NLS). The two homologous zinc finger proteins (Zn1 and Zn2) are characterized by a CCHC ligand pattern.32, 33 (2) The auto modification domain has BRCA1 C terminus (BRCT) motif and it is involved in protein‐protein interaction.2, 10, 34 (3) The catalytic domain at C terminus comprises of PARP signature motif (six β‐strands and one α‐helix) that binds to NAD+and glutamate residue at its 988 position.2
Figure 1.

Structural organization of human PARP‐1 (hPARP‐1): It is characterized by FI, FII: Zinc finger motifs, FIII: Zinc ribbon domain (1‐333 aa); NLS: Nuclear localization sequence; BRCT: BRCA1 C terminal motif (386‐464 aa); WGR domain (549‐634 aa) and the most conserved catalytic domain with PARP signature motif (PSM) between 859‐908 aa and Glutamate (Glu) at 988 position.
The next important component of this enzyme is the PARP signature motif (PSM). It has two sites, acceptor site for adenosine and donor site for nicotinamide wherein ADP residues from NAD+are transferred to target site.35 His‐862 and Glu‐988 play important role in NAD+binding.36 In addition to this, WGR domain also contains highly conserved amino acid sequence i.e. Trp, Gly and Arg, but its role is yet to be identified.2, 34 However, Langelier et al.37 showed that Zn3 along with Zn1 and WGR domain of PARP‐1 together bind to the DNA damage leading to structural changes eventually abridging DNA damage site to its catalytic domain.
Other than auto modification by PARylation, PARP‐1 itself undergoes various other modifications enlisted in Table 2 that has various cellular effects.38, 39, 40, 41, 42, 43
Table 2.
Post‐translational Modifications of Poly (ADP‐ribose) polymerase 1
| Modification in PARP‐1 | Source | Residue modified | Activator | Result | References |
|---|---|---|---|---|---|
| Auto modification Poly (ADP‐ribosylation) | PARP | K498, K521 and K524 | Intact and damaged DNA | Regulation of PARP activity | Altmeyer et al.38 |
| Mono‐ADP‐ribosylation | SIRT6 | K521 | dsDNA damage | Enhances double‐strand break repair under oxidative stress | Mao et al.42 |
| Sumolyation small ubiquitin‐related modifier (SUMO) | SUMO‐2SUMO 3 | K203, K486 andK512 | Heat shock intact DNA | Transcriptional co‐activator of hypoxia‐responsive genes and promotes induction of the heat shock‐induced HSP70.1 promoter | Zilio et al.43 |
| Acetylation | p300/CREB‐binding protein | K498, K505, K508, K521 and K524 | Inflammatory stimuli | NF‐κB‐dependent gene activation | Hassa et al.40 |
| Phosphorylation | ERK1/2Protein Kinase C | S372 and T373 | DNA damage | Neuronal cell deathDecreased PARP‐1 DNA‐binding and catalytic activity | Kauppinen et al.,41 Beckert et al.39 |
4.2. Mechanism of PARP‐1 activation
The enzymatic activity of PARP‐1 is stimulated significantly in the presence of a range of activators like damaged DNA, non‐B‐DNA structures, nucleosomes and various protein‐binding partners.1, 44, 45, 46 Lonskaya et al.47 reported that DNA bent, cruciform DNA or stably unpaired DNA regions can also stimulate PARylation. The activation signal for PARP‐1 is DNA damage although several reports illustrate that PARP‐1 may also be activated in the absence of DNA damage. The best characterized ligands for PARP‐1 are single‐strand and double‐strand breaks (SSBs and DSBs).
There are reports suggesting that PARP‐1 activation by SSBs requires presence of both the zinc fingers while only Zn1 is required for DSBs.33 Zn1 has been demonstrated to relay the signals to the catalytic domain for formation of PAR molecules,33, 48 while Zn2 has been shown to be majorly involved in DNA binding as compared to Zn1 due to its higher affinity to DNA.49 Eustermann et al.50 have demonstrated very recently how the two zinc fingers recognize SSBs and coordinate domain folding in PARP‐1 to control the activity of the C‐terminal catalytic domain. PARP‐1 has been reported to have affinity for intact DNA structures and recognizes specific octamer motif “RNNWCAAA” found in various gene promoters.51
Another mode of alternative DNA‐independent mode of PARP‐1 activation is based on kinase cascades. Phosphorylated ERK2 has been shown to significantly enhance and maximize PARP‐1 catalytic activity in the presence and absence of damaged DNA.41, 52 Interaction between PARP‐1 and a pre‐phosphorylated kinase has also been shown to mediate PARP‐1 activation.52, 53 Likewise, phosphorylation by activated calcium‐dependent protein kinase (CaMKII) is also capable of activating PARP‐1 enzyme during neuronal development thereby promoting the nuclear export of its negative regulator KIF4.54 Moreover, overexpression of protein phosphatase 5 (PP5) led to increase in PARP‐1 enzymatic activity in response to double‐stranded DNA breaks.55 Nuclear nicotinamide mononucleotide adenylyl transferase 1 (NMNAT1), an enzyme involved in NAD+synthesis, also associates with PAR to enhance PARP‐1 enzyme activity.56 Other proteins regulating PARP‐1 activity include Ku,55 histone variant macroH2A1.157 and KIF4.58 Protein‐protein interactions also seem to activate PARP‐1. Mao et al.42 have demonstrated that SIRT6 activates PARP‐1 by mono‐ADP‐ribosylating it in position Lys521. Developmental or environmental stimuli induce PARP‐1 activation and the PAR‐dependent nucleosome loosening leading to histone stripping and hence opening of the chromatin structure. This process allows transcriptional activation. This PAR‐mediated chromatin loosening phenomenon is detected at larval salivary‐gland polytene chromosome puffs.59 Hence, PARP‐1 can be activated by DNA‐dependent and ‐independent manner which is summarized in Fig 2.
Figure 2.

Mechanism of PARP‐1 Activation. The nuclear enzyme PARP‐1 can bind to DNA breaks resulting in the activation of the enzyme. DNA breaks are caused either by ROS, RNS or radiation or indirectly by DNA repair machinery where breaks are introduced into the DNA strands as in the case of alkylating DNA damage. Binding to special non‐B‐DNA structures such as bent or cruciform DNA or four‐way junctions may culminate into PARP‐1 activation. Protein‐protein interactions or covalent modifications (e.g. mono‐ADP‐ribosylation, acetylation or phosphorylation) have also been described as activation mechanisms for PARP‐1 which are DNA‐independent. Other proteins activating PARP include nuclear NMNAT, Ku and phosphorylated ERK2 and Histone‐4 tail
4.3. PARP‐1: single protein with varied roles
4.3.1. PARP‐1 in DNA repair
ADP‐ribosylation activity of PARP‐1 is an instantaneous biochemical response to DNA damage induced by ionizing radiations, alkylations etc. At low levels of DNA damage, it detects DNA damage followed by repair and cell survival, whereas at high levels of DNA damage, it activates the cell death pathway.60 Upon DNA damage, PARP‐1's zinc finger FI/Zn1, FII/Zn2 and FIII/Zn3 motifs have been reported to relay binding signal to catalytic domain followed by the recruitment of proteins involved in repair mechanism such as base excision repair (BER), single‐strand breaks (SSBs) and double‐strand breaks (DSBs) repair.1, 61 It is also indicated to act as a DNA damage sensor 62 and help in chromatin remodelling at DNA damage sites. 63 A variety of proteins like ALC1, histone mH2A1.1, scaffold attachment factor SAFB1 have been illustrated to be recruited to DNA damage sites via PARP‐1 thus proving its indispensable role in DNA repair.64, 65, 66 Evidences show presence of PAR‐binding zinc finger motifs in DNA damage response and checkpoint regulation proteins.67, 68 PARP‐2 was also shown to be involved in the later steps of BER/single‐strand break repair.69 In nucleotide excision repair, PARP‐1 inhibition or depletion has also shown to cause low efficiency of removal of UV‐induced DNA damage.70 Among mammalian DNA repair pathways, PARP‐1 has been also implicated in homologous recombination71 and non‐homologous end‐joining pathways.72 PARP‐1 has been reported to interact with replication fork protein (Timeless) in a PAR‐independent manner thereby allowing its recruitment to DSB sites to promote homologous recombination.73 Thus, it is clearly illustrated that PARP‐1 plays a vital role in DNA damage response.
4.3.2. PARP‐1 in cell death
Under normal physiological conditions, cell morphology, numbers, pattern and injury are taken care of by the process of apoptosis.74 The mode of cell death depends on the extent of DNA damage. Low DNA damage can activate PARP‐1 resulting in cell survival via DNA repair mechanisms. At moderate levels of DNA damage, cell undergoes apoptosis and PARP‐1 activation results into cleavage of PARP‐1 by caspases‐3 and ‐7 into two fragments (89 kDa and 24 kDa)75 which is believed to be a key feature of apoptosis.76 N‐terminal 24 kDa fragment remains in nucleolus and other 89 kDa fragment translocates from nucleus to cytosol wherein it acts as a target for autoimmunity.77 Severe DNA damage leads to programmed necrotic cell death through over‐activation of PARP‐1.78 Ring finger protein 146 (RNF146), a cytoplasmic E3‐ubiquitin ligase, acts as a direct interactor of PARP‐1 during this process and elicits release of PARP‐1 from the nucleus. This has been demonstrated during myocardial ischaemia‐reperfusion injury.79 On the other side, in caspase‐independent cell death, it plays an important role in the release of apoptosis‐inducing factor (AIF) from mitochondria to nucleus. Yu et al.80 have studied the dependence of PARP‐1 and AIF in caspase‐independent cell death which is termed as ‘parthanatos’. PARP‐1 has been reported to play a very crucial role in initiation and regulation of this type of cell death.81 Parthanatos has been detected in many disease conditions like stroke, Parkinsons, diabetes, etc.82 Upon PARP‐1 activation stimulated with various DNA‐damaging agents like NMDA, H2O2, etc., AIF translocates from mitochondria to nucleus and finally culminates into cell death.83, 84, 85 On the contrary, Mir et al. showed that staurosporine‐induced cell death did not involve PARP.86
PARP‐1 is also reported to be involved in autophagy induced by DNA damage.87 PARP‐1 via autophagy displays a cytoprotective role in oxidative stress‐induced necrotic cell death.88 Moreover, Son et al. 89 have also reported that cadmium‐mediated ROS generation leads to PARP‐1 activation and energy (ATP) reduction, eventually culminating into autophagy in skin epidermal cells. Wyrsch et al.90 have found that PARP‐1 and PARP‐2 control cytosolic Ca2+shifts from extracellular and intracellular sources during oxidative stress. The different Ca2+signals arise from the transient receptor potential melastatin 2 (TRPM2) channels located in the cellular and lysosomal membranes. This Ca2+overload induces specific stress kinase response which leads to autophagy or cell death. Under mild oxidative stress conditions, PARP‐1 operates as an autophagy suppressor after oxidative stress leading to cell death by activating downstream of extracellular signal‐regulated kinase 1/2 (ERK1/2) and AKT. Under severe oxidative conditions, PARP‐2 induces Ca2+shifts from lysosomes, while PARP‐1 becomes completely inactive. The cytosolic Ca2+overload leads to phosphorylation of p38, stress‐activated protein kinase/Jun amino‐terminal kinase (SAPK/JNK), and cyclic AMP response element‐binding protein (CREB) with its activating transcription factor (ATF‐1), further activating autophagy markers leading to cell survival.
PARP‐1 and related PARP family members are at the intersection of conversing stress signalling pathways. Oxidative stress causes disruption in redox potential that extends to the ER, causing accumulation of misfolded proteins, finally stimulating the unfolded protein response (UPR).91 It would be interesting to know if PARP‐1 has a role in ER stress‐mediated cell death as it is upstream to autophagy, where PARP‐1 is demonstrated to play an essential role. Hence, it is clear that PARP‐1 is an essential regulator in many of the cell death pathways and this has been demonstrated in many tissues. However, a very interesting work by Jog and Caricchio92 illustrates a characteristic difference in PARP‐1‐mediated necrosis in males and females. Male mice were shown to be prone to PARP‐1‐mediated necrosis while female mice showed PARP‐1‐independent cell death.92 Understanding the role of PARP‐1 in different stress conditions and even in different sexes would help us dissect out pathomechanisms of various disease conditions.
4.3.3. PARP‐1 and epigenetics
The poly (ADP‐ribosyl)ation of histones leading to open chromatin conformation at DNA damage sites was the first indication to the function of PAR as an epigenetic modification.2 Recent evidence has shown that PAR has an important role in the epigenetic regulation of chromatin structure and in gene expression under physiological conditions wherein DNA integrity is maintained.93 Lodhi et al.94 have demonstrated PARP‐1 as a genome‐wide epigenetic memory mark in mitotic chromatin. They report that PARP‐1 establishes stable epigenetic marks at the transcription start sites in metaphase chromatin and these marks are a prerequisite for transcriptional restart after mitosis. Moreover, PARP‐1 activity epigenetically regulates mitochondrial DNA repair and transcription.95 PARP‐1 also associates with genome‐wide epigenetic regulatory elements suggesting a functional interplay between PARP‐1 and DNA methylation.96 Previous studies have shown that PARP‐1 can affect the genomic DNA methylation pattern via DNA methyl transferase, Dnmt1, both by regulating its expression as well as activity.93, 97 Furthermore, the role of PARP‐1 in DNA methylation events has been explored in induced pluripotent stem cells (iPSCs).98 Recently, PARP‐1 has been shown to be associated epigenetically with Tet2 (a methyl cytosine dioxygenase) during somatic cell reprogramming which leads to transcriptional induction at the pluripotency loci.99 PARP‐1 has also been demonstrated to interact with TIP5 via non‐coding RNA, thereby playing a role in maintenance of silent rDNA chromatin in mid‐late S phase.100 Though, these studies suggest the possible epigenetic involvement of PARP‐1; its mechanistic role in epigenetic control is still elusive and remains to be an area of great interest to researchers.
4.3.4. PARP‐1 as a chromatin modulator
Chromatin consists of genomic DNA, linker histones (H1), core histones (H2A, H2B, H3 and H4) and other chromatin‐associated proteins. Early reports have shown that purified PARP‐1 could ADP‐ribosylate chromatin proteins (e.g. mainly H1), by de‐condensation of chromatin and destabilization of nucleosomes.101 Also proven in recent reports, PARP‐1 binding to chromatin can change the conformation and composition of nucleosome.32, 102 In addition, it has also been demonstrated that PARP‐1 interacts with core histone variants resulting in the recruitment and integration of histone variants to specific sites in the genome.57 Local chromatin loosening by PARP‐1 has also been demonstrated well at the puff loci in Drosophila facilitating transcription and eventually helping chromatin remodelling during development.59 Nalabothula et al.96 discussed the possible mechanisms of chromatin structure remodelling by PARP‐1 as: a) it binds between entry and exit sites between nucleosomes and linker DNA, b) it PARylates histones, linker histone H1, etc. thus modifying chromatin architecture and c) it competes with histone H1 for nucleosome binding. All the above reports strengthen the role of PARP‐1 in chromatin remodelling.
4.3.5. PARP‐1 in transcription
It is well studied that PARP‐1 behaves as chromatin modifier at transcriptional level with a number of in vitro and in vivo experiments. Electrostatic repulsion between DNA and histones due to transfer of negatively charged PAR molecules onto accepter proteins promotes transcription by recruiting transcriptional machinery.103 PARP‐1 is observed to be more localized at the promoter regions of most actively transcribed genes.104 The transcriptional regulatory roles of PARP‐1 are manifested mainly through two processes, modulating chromatin structure and acting as a part of enhancer/promoter‐binding complexes. Based on the cell type, it can enhance transcription with co‐activators or inhibit transcription by repressors.105 Chromatin‐dependent gene expression is controlled by PARP‐1 interacting with histones at promoter.104 The type of histone modification (acetylation, phosphorylation and methylation) is very essential for interaction between PARP‐1 and DNA because it can add structural changes into histones.106 Phosphorylation of histone variant, H2Av, promotes activity of PARP‐1 in Drosophila at specific promoter regions.107 PARP‐1 is also found to be localized at DNA repair sites after binding to other histone variant, macroH2A.108 Also, macroH2A1‐stimulated H2B acetylation was seen in cancer progression which was PARP‐1‐dependent.109 Depletion of PARP‐1 activity resulted into ineffective loading of RNA polymerase II transcriptional machinery implying its role in gene regulation.110
Recent studies suggest that PARP‐1 functions as a co‐activator, which upregulates the transcription of Nrf2, promoting the interaction among Nrf2 and ARE (antioxidant response elements).111 Reduced expression of CCN2 was found in tubular epithelial cells of kidney upon knockdown of PARP‐1.112 In addition to this, PARP‐1 also functions as an insulator that organizes the genome into distinct regulatory units by controlling the effects of enhancers on promoters, or by preventing the spread of heterochromatin.113 In vivo and in vitro binding studies of PARP‐1 and transcription factor Yin Yang 1 (YY1) suggested that PARP‐1 plays a promoter regulatory role and inhibits the transcription of Cxcl12. In addition, changes in PARP‐1‐CTCF interactions due to serum shock induced recruitment of circadian loci to the lamina leading to transcriptional attenuation.114 PARP‐1 is also known to be acting as an exchange factor thereby controlling transcription. Recently, it has been demonstrated that PARP‐1 functions in remodelling of promoter‐associated nucleosomes by replacing H2A.Z by H2A from FOS promoter to allow transcriptional activation in response to ERK signalling.115 Thus, the underlying mechanism of PARP‐1‐mediated transcriptional regulation is very complex and extensive and hence more studies are required to explore the transcriptional role of PARP‐1.
4.3.6. PARP and spermatogenesis
Both PARP‐1 and PARP‐2 have been found to have a significant role in spermatogenesis.116 It has been observed that there is significant PARP expression during the earlier stages of spermatogenesis and its transcription declines during late stages of maturation.117, 118 The levels of PARP‐1, PARP‐2 and PARP‐9 were found to be increased in mature sperms as compared to immature sperms 116 and interestingly PARP‐1 was also found to be downregulated during the haploid stage of meiosis.119 The presence of PARG in the nuclei of rat primary spermatocytes also suggests that the levels of poly (ADP‐ribose) in these germ cells are highly regulated.117 Moreover, Meyer‐Ficca et al.120 reported the presence of PAR polymerization by PARP‐1 and PARP‐2 in rat spermatids, highest during the phase of chromatin condensation.
Studies demonstrating an increase in DNA strand breaks in all population of elongating spermatids in human testis 121 and the presence of higher levels of PARP‐1, PARP‐2 and PARP‐9 in ejaculated sperm from fertile men compared to infertile men indicate a possible relationship between PARP expression and male infertility.
4.3.7. PARP‐1 in cell differentiation/multicellularity
Out of the various roles of PARP‐1, its role in cell differentiation and multicellularity has yet to be unravelled. However, accumulating reports in different model systems suggest a definite role of PARP‐1 in growth and multicellularity. For example, Drosophila PARP has been shown to act in ectodermal specification and neural crest development in zebrafish.122 Our laboratory studies are indicative of PARP's role in D.discoideum development wherein its downregulation led to arrested development.123 Recent studies from our laboratory show PARP‐1 involvement in D. discoideum growth and multicellularity by ADPRT1A (PARP‐1 orthologue) overexpression which led to delayed growth and developmental morphogenesis.124 We have also reported that PARP may be essential in combating stress conditions in D. discoideum.83, 84, 85, 125, 126 Genetic studies on PARP‐1 orthologues in fungus demonstrated defective development and decreased life span.127, 128, 129 As we move to the higher life forms like plants, it was seen that AtPARP‐1 and/or AtPARP2 knockdown reported to alter Arabidopsis development 130 and AtPARP2 orthologue in oilseed rape (Brassica napus) did not affect its development.131 However, further work is mandatory to explore the role of PARP in plant development. In addition, studies in Drosophila also suggest importance of PARP in chromatin loosening at ecdysone‐inducible regions thereby inducing purparium formation and metamorphosis.59, 132 These results are also substantiated by mice studies wherein PARP‐1 and PARP‐2 double‐mutant mice were found to be not viable and die at the onset of gastrulation, establishing the importance of both the PARPs during early embryogenesis.133 Recently, Hamazaki et al.134 have shown that PARP inhibition caused inhibition of DNA demethylation of the IL17d promoter region at the two‐cell stage leading to downregulation of genes essential for early embryogenesis. Thus it is clear from the above that a strong association of PARP‐1 exists in differentiation and multicellularity, which is yet to be explored in detail.
4.3.8. PARP‐1 in metabolic regulation
PARP‐1 has been known for its role in DNA repair as discussed in above sections. However, recent data suggest a role for PARP‐1 in metabolic regulation by influencing mitochondrial function and oxidative metabolism. Mouse knockout studies showed that PARP‐1 deletion led to increased food intake.135, 136 PARP‐1−/− mice showed an increased metabolic rate.137 PARP‐1 has also been associated with reduction in the glycolytic rate which has been linked to a reduction in NAD+availability over the years.138 Over‐activation of PARP activity can lead to metabolic perturbations through reduction in ATP, NAD+/NADH levels, which is enough to impair carbohydrate metabolism.139 It also changes the flow of glycolytic metabolites into Krebs cycle and thereby compromised energy production in mitochondria.140 However, recent evidence indicates that PARP‐1 may be responsible for reduction in hexokinase activity and hence affects the cellular glycolytic rate via poly (ADP‐ribosyl)ation of hexokinase directly.141 PARP‐1 and PARP‐2 activation have also been demonstrated to affect mitochondrial activity negatively.142 Hence, PARP inhibition arises as therapeutant to treat mitochondrial dysfunction.
In addition, PARP‐1 also plays a crucial role in the circadian entrainment and regulates feeding behaviour. Asher et al.143 demonstrated that CLOCK (Circadian transcription factor)–BMAL1‐dependent gene expression was altered in PARP‐1‐knockout mice, in response to changes in feeding times. In contrast, the deletion of PARP‐2 did not affect food intake or daily behaviour.135 Moreover, both PARP‐1−/− and PARP‐2−/− mice displayed enhanced energy expenditure.135, 138 PARP‐1−/− mice showed an increased mitochondrial content in their brown adipose tissue (BAT),138 which physiologically renders them to be able to maintain their body temperature during cold exposure. Interestingly, PARP‐2 deletion does not influence mitochondrial biogenesis in BAT.138 Furthermore, it has been suggested that PARP‐1 acts as a positive regulator of adipogenesis and adipocyte function resulting in fat deposition.144 Studies have confirmed that PARP‐1 regulates adipogenic gene expression and is required selectively for adipocyte function.145 PARP‐1−/− and PARP‐2−/− mice also displayed an increased glucose clearance 135, 138 suggesting the increased insulin sensitivity. Thus, these reports suggest the metabolic involvement of PARP‐1 and PARP‐2; however, more studies are needed to confirm these findings and to explore new metabolic regulatory functions of PARP.
4.3.9. PARP‐1 and cancer
Errors in replication process, production of ROS and UV radiations result in DNA damage which includes single‐strand breaks (SSBs), double‐strand breaks (DSBs), etc. Cells then signal DNA repair pathways such as nucleic acid excision repair (NER), base excision repair (BER), mismatch repair (MMR), non‐homologous end‐joining (NHEJ) and homologous recombination (HR) resulting into cell survival with an exception of tumour cells. PARP‐1 and PARP‐2 are key regulators for the function of DNA repair mechanisms; however, genetic disorders, such as BRCA1 and BRCA2 mutations, prevent DNA repair mechanism and increase the risk of malignancies.146 Inhibition of DNA repair process may lead to cell death and this brings PARP‐1 as a perfect target for anti‐cancer therapy. PARylation of targeted proteins by PARP‐1 on activation by SSBs and DSBs facilitates the recruitment of DNA repair proteins such as XRCC1 to sites of damage.147, 148 PARP‐1 may also facilitate HR via recruitment of factors like ataxia telangiectasia‐mutated (ATM, Ataxia Telangiectasia Mutated), Nijmegen breakage syndrome 1 (Nbs1) and mitotic recombination 11 (Mre11) to sites of DSBs.149 However, major role in HR repair involves localization of BRCA‐1 and BRCA‐2. BRCA‐1 plays an essential role in the surveillance of DNA damage and transduction of DNA repair responses, while BRCA‐2 is directly involved in double‐stranded DNA repair, via modulation of Rad51 by HR.150
PARP‐1 inhibition does not cause cell lethality by itself, as the cell has an intact HR pathway for DNA repair. Cells that have a mutated BRCA1 or BRCA2 genes as in the case of breast cancer or those that are deficient in BRCA1 or BRCA2 proteins like sporadic cancers are found to be defective in their ability to repair DNA through HR and henceforth depend on error‐prone NHEJ. This results in amplification of DNA instability and chromosomal aberrations eventually causing cell death (Fig 3a). This synergistic effect has been very well demonstrated by Arun et al.,151 wherein PARPi AZD2281 showed more promising results in BRCA1‐ and BRCA2‐bearing mutants via induction of autophagy. This concept of synthetic lethality has been implemented upon in cancer therapeutics. In cases of breast and ovarian cancer, treatment with PARP‐1 inhibitors Olaparib and Veliparib (Approach A) has found positive clinical results.152 Epigenetic modulation or artificial inactivation of BRCA pathway (Approach 2a) in cases of sporadic cancer along with the use of PARPi plays a key to therapeutics. This synergistic inhibition of DNA repair poses as a double‐hit mechanism for cancer cell death. PARPi can also be used in combination with chemotherapy and radiation (Approach 2b) to render the cells prone to cell death under enhanced damaged conditions as in cases of non‐Hodgkin lymphoma cell line, use of PARPi in combination with both external beam radiation and 131I‐tositumomab; radio sensitization with veliparib in head and neck carcinoma cell lines and lung cancer xenograft models; or with niraparib in neuroblastoma cell lines, and whole brain radiation in cases of brain metastases 153 (Fig 3b). In addition, Table 3 compiles various drug combinations with Olaparib (Table 3a) and Veliparib (Table 3b) which are being currently extensively used in various cancers along with its side effects.
Figure 3.

PARP‐1 and cancer therapy. (a) In normal cells, upon DNA damage like SSB, DSB and non‐B‐DNA structures, PARP‐1 gets activated and thereby aids in the recruitment of DNA repair proteins such as the scaffolding protein XRCC1 to sites of SSBs through BER, whereas DNA‐PKcs, Ku70 and Ku80 to sites of DSBs through NHEJ. It also aids HR via recruitment of factors like ATM, Mre11 and Nbs1 to sites of DSBs. Another very essential process of HR repair involves localization of BRCA‐1 and BRCA‐2 to sites of double‐stranded DNA damage. In cancer cells bearing BRCA1/2 mutations or deficiency (red star), cells are rendered faulty in HR repair (red no symbol) and thus there is complete dependence on NHEJ (error‐prone) for DSB DNA repair and SSB for BER (red arrows); both of which are PARP‐1‐dependent. Thus, PARP inhibition serves as an excellent approach for therapy. BRCA1/2 mutations or deficiency along with PARP‐1 inhibition leads to amplification of DNA instability due to impairment in BER‐, NHEJ‐ and ATM‐mediated HR repair and chromosomal aberrations results in cell death. (b) PARP‐1 inhibitors like Olaparib, Veliparib, etc. have been promising therapeutic candidates in case of breast cancer and ovarian cancer—Approach 1. Approach 2a uses PARP‐1 inhibitors in case of epigenetic modulation or artificial inactivation of BRCA pathway in case of sporadic cancers, whereas approach 2b involves use of chemotherapy and radiation along with PARP‐1 inhibitor depending on the cancer type
Table 3.
PARP inhibitors—Olaparib and Veliparib in various disorders
| Mode of administration | Disease/disorder | Nature of disorder | Clinical trial status and results | Dosage (mg) | Side effects/toxicity | Reference |
|---|---|---|---|---|---|---|
| (a) PARP inhibitor Olaparib in various cancers | ||||||
| Oral, monotherapy | Ovarian cancer | Recurrent; with BRCA1 or BRCA2 mutation | Phase II; positive | 200; Twice a day | Anaemia and Vomiting | Kaye et al.,167 Mateo et al.168 |
| 300; Twice a day | Anaemia | Mateo et al.168 | ||||
| 400; Twice a day | Fatigue, nausea, vomiting and decreased appetite | Gelmon et al.,169 Kaye et al.,167 Ledermann et al.,170 Kaufman et al.171 | ||||
| Advanced; without BRCA1/2 mutation | Phase II; negative | 400; Twice a day | — | Gelmon et al.169 | ||
| Triple‐negative breast cancer | Advanced; with BRCA1 or BRCA2 mutation | Phase II; positive | 100; Twice a day | Fatigue and nausea | Tutt et al.172 | |
| Breast cancer | Advanced; with BRCA1 or BRCA2 mutation | Phase II; positive | 400; Twice a day | Fatigue, nausea and vomiting | Tutt et al.172 | |
| Oral, Combination therapy with Cediranib | Ovarian cancer | Recurrent; with BRCA1 or BRCA2 mutation | Phase I; positive | 200/400 mg twice daily plus Cediranib 30 mg daily | Bowel obstruction, fatigue, hypertension, thrombocytopenia | Liu et al.173 |
| Oral, Combination therapy with Cediranib | Ovarian fallopian tube, or primary peritoneal cancer | Recurrent; platinum‐sensitive, or with BRCA1/2 mutations | Phase II; Positive compared to monotherapy (400 mg) | 200 mg twice daily plus Cediranib 30 mg daily vs 400 mg Olaparib | Fatigue, diarrhoea and hypertension | Liu et al.174 |
| Oral, Combination therapy with Cediranib | Triple‐negative breast cancer | Metastatic | Phase I; Negative | 200/400 mg twice daily plus Cediranib 30 mg daily | — | Liu et al.173 |
| Oral, Triple combination therapy with carboplatin and paclitaxel | Ovarian cancer | Advanced, with or w/o BRCA1 or BRCA2 mutations, relapsed | Phase Ib/II; Positive | 200 mg twice daily plus carboplatin and paclitaxel, followed by olaparib 400 mg | Neutropenia and anaemia | Oza et al.,175 Rivkin et al.176 |
| Oral, Triple combination therapy with carboplatin+paclitaxel and Chemo therapy | Ovarian cancer | Advanced, with or without BRCA1 or BRCA2 mutations, relapsed | Phase I; Positive | 200 mg twice daily plus carboplatin and paclitaxel, followed by olaparib 400 mg | Alopecia, nausea, neutropenia, diarrhoea, headache, peripheral neuropathy and dyspepsia | Oza et al.175 |
| Oral; with Chemo therapy | Ewing sarcoma | Refractory | Phase II; Negative | 400; Twice a day | Anaemia and thrombocytopenia | Choy et al.177 |
| Oral, monotherapy | Pancreatic cancer | With germline BRCA1/2 mutation | Phase II; Positive | 400; Twice a day | Fatigue, nausea, anaemia and vomiting | Kaufman et al.171 |
| Oral, monotherapy | Prostate cancer | With germline BRCA1/2 mutation | Phase II; Positive | 400; Twice a day | Fatigue, nausea, anaemia and vomiting | Kaufman et al.171 |
| Intravenous; Combined with cetuximab (cet) and intensity modulated radiation therapy | Head and neck squamous cell carcinoma | Heavy smokers, locally advanced | Phase I; Uncertain/ongoing | 400 mg/m2+cet 250 mg/m2 IV. | Mucositis, dermatitis, clinically insignificant lymphopenia, and hypomagnesaemia | Waxweiler et al.178 |
| Oral; with temozolomide | Glioblastoma | Relapsed | Phase I; Positive penetration | 400 | — | Chalmers et al.179 |
| Oral; with topotecan | Advanced solid tumours | — | Phase I; Negative | 100 mg twice daily | — events | Samol et al.180 |
| (b) PARP inhibitor Veliparib in various cancers | ||||||
| Oral, monotherapy | Epithelial ovarian cancer | Recurrent or persistent; With germline BRCA1/2 mutation | Phase II; Negative; discontinued | 400 | Grade 3‐fatigue, nausea, leukopenia, neutropenia, dehydration, and ALT. Grade 2 events‐nausea, fatigue, vomiting and anaemia | Coleman et al.181 |
| Oral; monotherapy | Serous ovarian cancer | Without BRCA1/2 mutation | Phase I; Negative | 400 | Nausea/vomiting, fatigue and leukopenia | Pahuja et al.182 |
| Oral; monotherapy | Triple‐negative breast cancer | Without BRCA1/2 mutation | Phase I; Negative | 400 | — | Pahuja et al.182 |
| Oral; monotherapy | Castration‐resistant prostate cancer | BRCA2‐mutated metastatic | Phase I; Positive | 400 | — | Pahuja et al.182 |
| Oral; with irinotecan | Triple‐negative breast cancer | Without BRCA1/2 mutation | Phase I; Negative | 40 mg+irinotecan 100 mg/m2 | Leukopenia, neutropenia, nausea, diarrhoea, fatigue, anaemia and vomiting | LoRusso et al.183 |
| With BRCA1/2 mutation | Phase I; positive | |||||
| Oral; Combined with Cisplatin and etoposide | Small cell lung cancer | Previously untreated | Phase I; positive | 100+Cisplatin 75 mg/m2+etoposide 100 mg/m2 | Dehydration, diarrhoea, fatigue, febrile neutropenia, heart failure, leukopenia, lymphopenia, nausea, neutropenia, respiratory failure and thrombocytopenia | Owonikoko et al.184 |
| Oral, Combined with carboplatin and paclitaxel | Squamous (Sq) non‐small cell lung cancer | Untreated advanced/metastatic | Phase III; | 120 mg+carboplatin AUC 6 mg/mL/m IV and paclitaxel 200 mg/m2 IV | — | McKee et al.185 |
| Oral; with metronomic Cyclophosphamide | Solid tumours and lymphomas | Advanced | Phase I; Positive | 60 mg+cyclophosphamide 50 mg | — | Kummar et al.186 |
| Oral; with cisplatin and gemcitabine | Pancreas adenocarcinoma | Potential BRCA/PABL2 mutated | Phase Ib; Positive | 80 mg+C 25 mg/m2 IV, G 600 mg/m2 IV | Anaemia, neutropenia, thrombocytopenia, haematologic toxicity and fatigue | O'Reilly et al.187 |
| Oral; With Temozolomide (TMZ) | Castration‐resistant prostate cancer | Docetaxel‐pretreated patients with metastatic | Phase I; positive | 40 mg+TMZ 150 mg/m2 | Thrombocytopenia, anaemia, fatigue, neutropenia, nausea and constipation | Hussain et al.188 |
The transcriptional role of PARP‐1 in cancer includes chromatin modulation of tumour suppressor and oncogene function, regulation of the metastatic processes, alteration of cell survival and adaptation. For example, in liver cancer, ATPases activity of ALC1 (amplified in liver cancer 1) was found to be dependent upon both PARP‐1 and NAD+. 154 Furthermore, various tumour cell lines exhibited overexpression of PARP‐1 with malignancy progression.155 One of the recent studies indicated that following irradiation, PARP‐1 activation plays a critical role in prostate cancer cell lines (LNCaP and DU145).156
PARP‐1 is also thought to be an important modulator of tumour suppressor gene, p53.157 In addition, PARP‐1 is known to regulate organ site‐specific tumour suppressors as explained by tumour suppressor gene, APC (adenomatous polyposis coli). The loss of this gene was associated with sporadic colorectal cancer (CRC). Collective reports suggest that PARP‐1 controls activity of T‐cell factor (TCF)/lymphoid enhancer factor (LEF), i.e. TCF/LEF complex in CRC with higher expression levels of PARP‐1.158, 159 In addition, Schiewer et al. 160 showed that PARP‐1 controls androgen receptor (AR) association and functions with chromatin using in vitro and in vivo systems. In particular, reduction in AR activity was correlated with significant anti‐tumour response to PARP‐1 inhibition, indicating the dependence of prostate cancer on PARP‐1 activity.160 Thus, these studies suggest that inhibition of PARP‐1 has potential as a cancer therapeutic through at least two mechanisms: (1) by potentiating chemotherapeutic agents that damage DNA and increasing tumour sensitivity; and (2) by inducing “synthetic lethality” in cells that are highly dependent on PARP‐1, due to deficiency in homologous recombination such as BRCA1 mutants.
4.3.10. Clinical implications of PARP‐1 in other diseases
Dysfunctional PARP‐1 has been linked to the onset and progression of myriad of diseases including cancer, ageing, diabetes, neurological diseases, etc. Several evidences point out the role of PARP‐1 in cancer. In addition, PARP‐1 has also been associated in neuronal pathology. PARP‐1 inhibition has been proven to play a protective role in Parkinsons and Alzheimer's disease.161 Moroni et al. also illustrated PARP‐1 inhibitor HYDAMTIQ to be very effective in conferring neuroprotection post stroke.162 In addition, PARP‐1 activation plays a role in diabetic nephropathy, neuropathy and retinopathy. Studies in experimental models reflect the role of PARP‐1 in inflammatory responses by promoting inflammation‐relevant gene expression. Moreover, activation of NF‐κB, AP‐1 and heat shock factor protein‐1 transcription factors, classically known to signal inflammatory gene expression are mediated by PARP‐1.163, 164 PARP‐1 also controls immunosuppressive function of regulatory T cells by destabilizing Foxp3.165 Also, an increase in Foxp3+T regulatory cells has been observed in PARP‐1 deficiency.166 PARP‐1 has thus emerged as a very important therapeutic target not only in cancer but also in several other diseases which can be further probed for its therapeutic potential.
5. Conclusion
The current research in PARP‐1 biology unravels the role of PARP‐1 beyond DNA repair and its involvement in several biological/cellular processes, such as epigenetics, transcriptional regulation, spermatogenesis, differentiation, etc. (Fig 4). The role of PARP‐1 as a transcriptional regulator has shed light on the broader aspect of PARP‐1 in the cell. Recent studies have also highlighted the multifaceted role of PARP‐1 in transcriptional regulation and provided new insights into how PARP‐1 plays a very important role in signalling pathways in the cell. In addition, PARP‐1's potential in therapeutics for diverse disease conditions require more animal‐based clinical studies. Much work needs to be done to understand how PARP‐1 works in conjunction with the other PARP family members. Moreover, PARP‐1 inhibitors have been a promising therapeutic for a wide range of pathological conditions. Inhibiting PARP activity uncovers potential of PARP inhibitors as promising candidates for cancer therapy, particularly in BRCA1/2‐mutated cancers, alone or in combination with cytotoxic drugs. p53‐deficient breast cancer cells treated with a PARP inhibitor happen to lose resistance to an apoptosis promoting, clinically active anti‐tumour agent called doxorubicin. However, these PARP inhibitors have several side effects that are toxic to the cell as the reports clearly show PARP‐1's role in physiological conditions. Hence, to harness the therapeutic potential of PARP‐1, studies are required to find out new inhibitors with least side effects. Thus, PARP‐1 has now opened new avenues for researchers to understand PARP‐1's multifunctional role in the cell which would eventually aid to further expand the utility of PARP family and its inhibition in therapeutics.
Figure 4.

PARP‐multifunctional protein. Poly ADP‐ribose polymerase family of proteins are detected from prokaryotes to eukaryotes. The number of PARP family members and their involvement in various cellular processes increases with the complexity of the organism. PARP‐1, the most studied PARP family member is involved a wide range of processes like DNA repair, cell death, transcription, spermatogenesis, metabolic regulation, epigenetics and differentiation
Acknowledgements
RB thanks Council of Scientific and Industrial Research, New Delhi (38 (1383)/14/EMR‐II) Department of Biotechnology, New Delhi (BT/PR4383/BRB/10/1014/2011) and Department of Science and Technology, New Delhi (SR/SO/BB‐03/2010) and for financial support. TJ thanks UGC‐SAP DRS for fellowship. RB thanks Dr. Mitesh Dwivedi for his critical suggestions. It is to be noted that none of the authors have any conflict of interest.
References
- 1. D'Amours D, Desnoyers S, D'Silva I, Poirier GG. Poly(ADP‐ribosyl)ation reactions in the regulation of nuclear functions. Biochem J. 1999;342:249–268. [PMC free article] [PubMed] [Google Scholar]
- 2. Schreiber V, Dantzer F, Ame JC, de Murcia G. Poly(ADP‐ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol. 2006;7:517–528. [DOI] [PubMed] [Google Scholar]
- 3. Lautier D, Lagueaux J, Thibodeau J, Ménard L, Poirier GG. Molecular and biochemical features of poly (ADP‐ribose) metabolism. Mol Cell Biochem. 1993;122:171–193. [DOI] [PubMed] [Google Scholar]
- 4. 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] [PubMed] [Google Scholar]
- 5. Min W, Wang ZQ. Poly (ADP‐ribose) glycohydrolase (PARG) and its therapeutic potential. Front Biosci (Landmark Ed). 2009;14:1619–1626. [DOI] [PubMed] [Google Scholar]
- 6. Oka S, Kato J, Moss J. Identification and Characterization of a Mammalian 39‐kDa Poly(ADP‐ribose) Glycohydrolase. JBC. 2006;281:705–713. [DOI] [PubMed] [Google Scholar]
- 7. Dunn CA, O'Handley SF, Frick DN, Bessman MJ. Studies on the ADP‐ribose pyrophosphatase subfamily of the nudix hydrolases and tentative identification of trgB, a gene associated with tellurite resistance. J Biol Chem. 1999;274:32318–32324. [DOI] [PubMed] [Google Scholar]
- 8. Rosenthal F, Feijs KLH, Frugier E, et al. Macrodomain‐containing proteins are new mono‐ADP‐ribosylhydrolases. Nat Struct Mol. 2013;20:502–509. [DOI] [PubMed] [Google Scholar]
- 9. Ame JC, Spenlehauer C, de Murcia G. The PARP superfamily. BioEssays. 2004;26:882–893. [DOI] [PubMed] [Google Scholar]
- 10. Hottiger MO, Hassa PO, Luscher B, Schuler H, Koch‐Nolte F. Toward a unified nomenclature for mammalian ADP‐ribosyltransferases. Trends Biochem Sci. 2010;35:208–219. [DOI] [PubMed] [Google Scholar]
- 11. Morales J, Li L, Fattah FJ, et al. Review of poly(ADP‐Ribose) polymerase (PARP) mechanisms of action and rationale for targeting in cancer and other diseases. Crit Rev Eukaryot Gene Expr. 2014;24:15–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Loseva O, Jemth AS, Bryant HE, et al. PARP‐3 is a mono‐ADP‐ribosylase that activates PARP‐1 in the absence of DNA. J Biol Chem. 2010;285:8054–8060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Hassa PO, Haenni SS, Elser M, Hottiger MO. Nuclear ADP‐ribosylation reactions in mammalian cells: where are we today and where are we going? Microbiol Mol Biol Rev. 2006;70:789–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Kickhoefer VA, Siva AC, Kedersha NL, et al. The 193‐kD vault protein, VPARP, is a novel poly(ADP‐ribose) polymerase. J Cell Biol. 1999;146:917–928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Smith S, Giriat I, Schmitt A, de Lange T. Tankyrase, a poly(ADP‐ribose) polymerase at human telomeres. Science. 1998;282:1484–1487. [DOI] [PubMed] [Google Scholar]
- 16. Seimiya H. The telomeric PARP, tankyrases, as targets for cancer therapy. Br J Cancer. 2006;94:341–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Leung AK, Vyas S, Rood JE, Bhutkar A, Sharp PA, Chang P. Poly(ADP‐Ribose) Regulates Stress Responses and MicroRNA Activity in the Cytoplasm. Mol Cell. 2011;42:489–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Ma Q, Baldwin KT, Renzelli AJ, McDaniel A, Dong L. TCDD‐inducible poly(ADP‐ribose) polymerase: a novel response to 2,3,7,8‐tetrachlorodibenzo‐p‐dioxin. Biochem Biophys Res Commun. 2001;289:499–506. [DOI] [PubMed] [Google Scholar]
- 19. Vyas S, Chesarone‐Cataldo M, Todorova T, Huang YH, Chang P. A systematic analysis of the PARP protein family identifies new functions critical for cell physiology. Nat Commun. 2013;4:2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Barkauskaite E, Jankevicius G, Ladurner AG, Ahel I, Timinszky G. The recognition and removal of cellular poly(ADP‐ribose) signals. FEBS J. 2013;280:3491–3507. [DOI] [PubMed] [Google Scholar]
- 21. Slade D, Dunstan MS, Barkauskaite E, et al. The structure and catalytic mechanism of a poly(ADP‐ribose) glycohydrolase. Nature. 2011;477:616–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Perina D, Mikoč A, Ahel J, Ćetković H, Žaja R, Ahel I. Distribution of protein poly(ADP‐ribosyl)ation systems across all domains of life. DNA Repair. 2014;23:4–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Faraone‐Mennella MR, Gambacorta A, Nicolaus B, Farina B. Purification and biochemical characterization of a poly(ADP‐ribose) polymerase‐like enzyme from the thermophilic archaeon Sulfolobus solfataricus. Biochem J. 1998;335:441–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Otto H, Reche PA, Bazan F, Dittmar K, Haag F, Koch‐Nolte F. In silico characterization of the family of PARP‐like poly(ADP‐ribosyl) transferases (pARTs). BMC Genom. 2005;6:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Li Z, Yamauchi Y, Kamakura M, et al. Herpes simplex virus requires poly(ADP‐ribose) polymerase activity for efficient replication and induces extracellular signal‐related kinase‐dependent phosphorylation and ICP0‐dependent nuclear localization of tankyrase 1. J Virol. 2012;86:492–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Mattiussi S, Tempera I, Matusali G, Mearini G, Lenti L, Fratarcangeli S. Inhibition of poly(ADP‐ribose)polymerase impairs Epstein Barr Virus lytic cycle progression. Infect Agent Cancer. 2007;2:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Ogura T, Takenouchi N, Yamaguchi M, Matsukage A, Sugimura T, Esumi H. Striking similarity of the distribution patterns of the poly(ADP‐ribose) polymerase and DNA polymerase beta among various mouse organs. Biochem Biophys Res Commun. 1990;172:377–384. [DOI] [PubMed] [Google Scholar]
- 28. Babiychuk E, Cottrill PB, Storozhenko S, Fuangthong M, Chen Y, O'Farrell MK. Higher plants possess two structurally different poly(ADP‐ribose) polymerases. Plant J. 1998;15:635–645. [DOI] [PubMed] [Google Scholar]
- 29. Citarelli M, Teotia S, Lamb RS. Evolutionary history of the poly(ADP‐ribose) polymerase gene family in eukaryotes. BMC Evol Biol. 2010;10:308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Kawal AM, Mir H, Ramniklal CK, Rajawat J, Begum R. Structural and evolutionary analysis of PARPs in D. discoideum . Am J Infect Dis. 2011;7:67–74. [Google Scholar]
- 31. Kameshita I, Matsuda Z, Taniguchi T, Shizuta Y. Poly(ADP‐ribose) synthetase. Separation and identification of three proteolytic fragments as the substrate‐binding domain, the DNA‐binding domain and the automodification domain. J Biol Chem. 1984;259:4770–4776. [PubMed] [Google Scholar]
- 32. Langelier MF, Ruhl DD, Planck JL, Kraus WL, Pascal JM. The Zn3 domain of human poly(ADP‐ribose) polymerase‐1 (PARP‐1) functions in both DNA‐dependent poly(ADP‐ribose) synthesis activity and chromatin compaction. J Biol Chem. 2010;285:18877–18887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Langelier MF, Servent KM, Rogers EE, Pascal JM. A third zinc‐binding domain of human poly(ADP‐ribose) polymerase‐1 coordinates DNA dependent enzyme activation. J Biol Chem. 2008;283:4105–4114. [DOI] [PubMed] [Google Scholar]
- 34. Ye'lamos J, Farres J, Llacuna L, Ampurdanes C, Martin‐Caballero J. PARP‐1 and PARP‐2: new players in tumour development. Am J Cancer Res. 2011;1:328–346. [PMC free article] [PubMed] [Google Scholar]
- 35. Kinoshita T, Nakanishi I, Warizaya M, et al. Inhibitor‐induced structural change of the active site of human poly(ADP‐ribose) polymerase. FEBS Lett. 2004;556:43–46. [DOI] [PubMed] [Google Scholar]
- 36. Masson MV, Rolli FD, Dantzer F, et al. Poly(ADP‐ribose) polymerase: structure‐function relationship. Biochimie. 1995;77:456–461. [DOI] [PubMed] [Google Scholar]
- 37. Langelier MF, Planck JL, Roy S, Pascal JM. Structural basis for DNA damage–dependent poly (ADP‐ribosyl)ation by human PARP‐1. Science. 2012;336:728–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Altmeyer M, Messner S, Hassa PO, Fey M, Hottiger MO. Molecular mechanism of poly(ADP‐ribosyl)ation by PARP1 and identification of lysine residues as ADP‐ribose acceptor sites. Nucleic Acids Res. 2009;37:3723–3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Beckert S, Farrahi F, Perween GQ, et al. IGF‐I‐induced VEGF expression in HUVEC involves phosphorylation and inhibition of poly(ADP‐ribose) polymerase. Biochem Biophys Res Commun. 2006;341:67–72. [DOI] [PubMed] [Google Scholar]
- 40. Hassa PO, Haenni SS, Buerki C, Meier NI, Lane WS, Owen H. Acetylation of poly(ADP‐ribose) polymerase‐1 by p300/CREB‐binding protein regulates coactivation of NF‐kappaB‐dependent transcription. J Biol Chem. 2005;280:40450–40464. [DOI] [PubMed] [Google Scholar]
- 41. Kauppinen TM, Chan WY, Suh SW, Wiggins AK, Huang EJ, Swanson RA. Direct phosphorylation and regulation of poly(ADP‐ribose) polymerase‐1 by extracellular signal regulated kinases 1/2. Proc Natl Acad Sci USA. 2006;103:7136–7141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Mao Z, Hine C, Tian X, et al. SIRT6 promotes DNA repair under stress by activating PARP‐1. Science. 2011;332:1443–1446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Zilio N, Williamson CT, Eustermann S, et al. DNA‐dependent SUMO modification of PARP‐1. DNA Repair. 2013;12:761–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kim MY, Zhang T, Kraus WL. Poly(ADP‐ribosyl)ation by PARP‐1: ‘PARlaying’ NAD+into a nuclear signal. Genes Dev. 2005;19:1951–1967. [DOI] [PubMed] [Google Scholar]
- 45. Kun E, Kirsten E, Ordahl CP. Coenzymatic activity of randomly broken or intact double‐stranded DNAs in auto and histone H1 trans‐poly(ADP‐ribosylation), catalyzed by poly(ADP‐ribose) polymerase (PARP I). J Biol Chem. 2002;277:39066–39069. [DOI] [PubMed] [Google Scholar]
- 46. Oei SL, Shi Y. Poly (ADP‐ribosyl)ation of transcription factor Y in Yang 1 under conditions of DNA damage. Biochem Biophys Res Commun. 2001;285:27–31. [DOI] [PubMed] [Google Scholar]
- 47. Lonskaya I, Potaman VN, Shlyakhtenko LS, Oussatcheva EA, Lyubchenko YL, Soldatenkov VA. Regulation of poly(ADP‐ribose) polymerase‐1 by DNA structure‐specific binding. J Biol Chem. 2005;280:17076–17083. [DOI] [PubMed] [Google Scholar]
- 48. Eustermann S, Videler H, Yang JC, et al. The DNA‐binding domain of human PARP‐1 interacts with DNA single‐strand breaks as a monomer through its second zinc finger. J Mol Biol. 2011;407:149–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Langelier MF, Planck JL, Roy S, Pascal JM. Crystal structures of poly (ADP‐ribose) polymerase‐1 (PARP‐1) zinc fingers bound to DNA structural and functional insights into DNA‐dependent PARP‐1 activity. J Biol Chem. 2011;286:10690–10701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Eustermann S, Wu WF, Langelier MF, et al. Structural Basis of Detection and Signaling of DNA Single‐Strand Breaks by Human PARP‐1. Mol Cell 2015;60:742–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Ko HL, Ren EC. Novel poly(ADP‐ribose) polymerase 1 binding motif in hepatitis B virus core promoter impairs DNA damage repair. Hepatology. 2011;54:1190–1198. [DOI] [PubMed] [Google Scholar]
- 52. Cohen‐Armon M, Visochek L, Rozensal D, et al. DNA‐independent PARP‐1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation. Mol Cell. 2007;25:297–308. [DOI] [PubMed] [Google Scholar]
- 53. Cohen‐Armon M. PARP‐1 activation in the ERK signaling pathway. Trends Pharmacol Sci. 2007;28:556–560. [DOI] [PubMed] [Google Scholar]
- 54. Ju B, Solum D, Song EJ, et al. Activating the PARP‐1 sensor component of the Groucho/TLE1 corepressor complex mediates a CaMKinase IIdelta‐dependent neurogenic gene activation pathway. Cell. 2004;119:815–829. [DOI] [PubMed] [Google Scholar]
- 55. Dong F, Soubeyrand S, Haché RJ. Activation of PARP‐1 in response to bleomycin depends on the Ku antigen and protein phosphatase 5. Oncogene. 2010;29:2093–2103. [DOI] [PubMed] [Google Scholar]
- 56. Zhang T, Berrocal JG, Yao J, DuMond ME, Krishnakumar R, Ruhl DD. Regulation of poly(ADP‐ribose) polymerase‐1‐dependent gene expression through promoter‐directed recruitment of a nuclear NAD+synthase. J Biol Chem. 2012;287:12405–12416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Ouararhni K, Hadj‐Slimane R, Ait‐Si‐Ali S, Robin P, Mietton F, Harel‐Bellan A. The histone variant mH2A1.1 interferes with transcription by down‐regulating PARP‐1 enzymatic activity. Genes Dev. 2006;20:3324–3336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Midorikawa R, Takei Y, Hirokawa N. KIF4 motor regulates activity‐dependent neuronal survival by suppressing PARP‐1 enzymatic activity. Cell. 2006;125:371–383. [DOI] [PubMed] [Google Scholar]
- 59. Tulin A, Spradling A. Chromatin loosening by poly(ADP‐ribose) polymerase (PARP) at Drosophila Puff Loci. Science. 2003;299:560–562. [DOI] [PubMed] [Google Scholar]
- 60. Virág L, Szabó C. The therapeutic potential of poly(ADP‐ribose) polymerase inhibitors. Pharmacol Rev. 2002;54:375–429. [DOI] [PubMed] [Google Scholar]
- 61. Masutani M, Nakagama H, Sugimura T. Poly(ADP‐ribose) and carcinogenesis. Genes Chromosom Cancer. 2003;38:339–348. [DOI] [PubMed] [Google Scholar]
- 62. Dantzer F, de La Rubia G, Menissier‐De MJ, Hostomsky Z, de Murcia G, Schreiber V. Base excision repair is impaired in mammalian cells lacking poly(ADP‐ribose) polymerase‐1. Biochemistry. 2000;39:7559–7569. [DOI] [PubMed] [Google Scholar]
- 63. Izhar L, Adamson B, Ciccia A, et al. A systematic analysis of factors localized to damaged chromatin reveals PARP‐dependent recruitment of transcription factors. Cell Rep. 2015;11:1486–1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Ahel D, Hořejší Z, Wiechens N, et al. Poly (ADP‐ribose) dependent regulation of DNA repair by the chromatin remodeling enzyme ALC1. Science. 2009;325:1240–1243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Altmeyer M, Toledo L, Gudjonsson T, et al. The chromatin scaffold protein SAFB1 renders chromatin permissive for DNA damage signaling. Mol Cell. 2013;52:206–220. [DOI] [PubMed] [Google Scholar]
- 66. Timinszky G, Till S, Hassa PO, et al. A macrodomain‐containing histone rearranges chromatin upon sensing PARP1 activation. Nat Struct Mol Biol. 2009;16:923–929. [DOI] [PubMed] [Google Scholar]
- 67. Ahel I, Ahel D, Matsusaka T, et al. Poly (ADP‐ribose)‐binding zinc finger motifs in DNA repair/checkpoint proteins. Nature. 2008;451:81–85. [DOI] [PubMed] [Google Scholar]
- 68. Rulten SL, Cortes‐Ledesma F, Guo L, Iles NJ, Caldecott KW. APLF (C2orf13) is a novel component of poly (ADP‐ribose) signaling in mammalian cells. Mol Cell Biol. 2008;28:4620–4628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Mortusewicz O, Amé JC, Schreiber V, Leonhardt H. Feedback‐regulated poly(ADP‐ribosyl)ation by PARP‐1 is required for rapid response to DNA damage in living cells. Nucleic Acids Res. 2007;35:7665–7675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Robu M, Shah RG, Petitclerc N, Brind'Amour J, Kandan‐Kulangara F, Shah GM. Role of poly(ADP‐ribose) polymerase‐1 in the removal of UV‐induced DNA lesions by nucleotide excision repair. Proc Natl Acad Sci USA 2013;110:1658–1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Adamson B, Smogorzewska A, Sigoillot FD, King RW, Elledge SJ. A genome‐wide homologous recombination screen identifies the RNA‐binding protein RBMX as a component of the DNA‐damage response. Nat Cell Biol. 2012;14:318–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Patel AG, Sarkaria JN, Kaufmann SH. Nonhomologous end joining drives poly(ADP‐ribose) polymerase (PARP) inhibitor lethality in homologous recombination‐deficient cells. Proc Natl Acad Sci USA. 2011;108:3406–3411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Xie S, Mortusewicz O, Ma HT, et al. Timeless interacts with PARP‐1 to promote homologous recombination repair. Mol Cell. 2015;60:163–167. [DOI] [PubMed] [Google Scholar]
- 74. Galluzzi L, Joza N, Tasdemir E, et al. No death without life: vital functions of apoptotic effectors. Cell Death Differ. 2008;15:1113–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Soldani C, Scovassi AI. Poly(ADP‐ribose) polymerase‐1 cleavage during apoptosis: an update. Apoptosis. 2002;7:321–328. [DOI] [PubMed] [Google Scholar]
- 76. Tewari M, Quan LT, O'Rourke K, et al. Yama/CPP32 beta, a mammalian homolog of CED‐3, is a CrmA inhibitable protease that cleaves the death substrate poly(ADP‐ribose) polymerase. Cell. 1995;81:801–809. [DOI] [PubMed] [Google Scholar]
- 77. Scovassi AI, Diederich M. Modulation of poly(ADP‐ribosylation) in apoptotic cells. Biochem Pharmacol. 2004;68:1041–1047. [DOI] [PubMed] [Google Scholar]
- 78. Sosna J, Voigt S, Mathieu S, et al. TNF‐induced necroptosis and PARP‐1‐mediated necrosis represent distinct routes to programmed necrotic cell death. Cell Mol Life Sci. 2014;71:331–348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Gerö D, Szoleczky P, Chatzianastasiou A, Papapetropoulos A, Szabo C. Modulation of Poly (ADP‐Ribose) Polymerase‐1 (PARP‐1)‐Mediated Oxidative Cell Injury by Ring Finger Protein 146 (RNF146) in Cardiac Myocytes. Mol Med. 2014;20:313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Yu SW, Andrabi SA, Wang H, et al. Apoptosis‐inducing factor mediates poly(ADP‐ribose) (PAR) polymer‐induced cell death. Proc Natl Acad Sci USA. 2006;103:18314–18319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Donizy P, Halon A, Surowiak P, Pietrzyk G, Kozyra C, Matkowski R. Correlation between PARP‐1 immunoreactivity and cytomorphological features of parthanatos, a specific cellular death in breast cancer cells. Eur J Histochem. 2013;57:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Andrabi SA, Dawson TM, Dawson VL. Mitochondrial and nuclear cross talk in cell death. Ann NY Acad Sci. 2008;1147:233–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Rajawat J, Alex T, Mir H, Kadam A, Begum R. Proteases involved during oxidative stress induced poly(ADP‐ribose) polymerase mediated cell death in D. discoideum . Microbiology. 2014;160:1101–1111. [DOI] [PubMed] [Google Scholar]
- 84. Rajawat J, Mir H, Alex T, Bakshi S, Begum R. Involvement of poly(ADP‐ribose) polymerase in paraptotic cell death of D. discoideum . Apoptosis. 2014;19:90–101. [DOI] [PubMed] [Google Scholar]
- 85. Rajawat J, Vohra I, Mir H, Gohel D, Begum R. Effect of oxidative stress and involvement of poly(ADP‐ribose) polymerase (PARP) in Dictyostelium discoideum development. FEBS J. 2007;274:5611–5618. [DOI] [PubMed] [Google Scholar]
- 86. Mir H, Rajawat J, Begum R. Staurosporine induced cell death in D. discoideum is independent of PARP. Indian J Exp Biol. 2012;50:80–86. [PubMed] [Google Scholar]
- 87. Muñoz‐Gámez JA, Rodríguez‐Vargas JM, Quiles‐Pérez R, et al. PARP‐1 is involved in autophagy induced by DNA damage. Autophagy. 2009;5:61–74. [DOI] [PubMed] [Google Scholar]
- 88. Huang Q, Shen HM. To die or to live: the dual role of poly(ADP‐ribose) polymerase‐1 in autophagy and necrosis under oxidative stress and DNA damage. Autophagy. 2009;5:273–276. [DOI] [PubMed] [Google Scholar]
- 89. Son YO, Wang X, Hitron JA, et al. Cadmium induces autophagy through ROS‐dependent activation of the LKB1–AMPK signaling in skin epidermal cells. Toxicol Appl Pharmacol. 2011;255:287–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Wyrsch P, Blenn C, Bader J, Althaus FR. Cell death and autophagy under oxidative stress: roles of poly(ADP‐Ribose) polymerases and Ca(2+). Mol Cell Biol. 2012;32:3541–3553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Mansuri MS, Singh M, Jadeja SD, et al. Could ER stress be a major link between oxidative stress and autoimmunity in Vitiligo? Pigmentary Disord. 2014;1:123. [Google Scholar]
- 92. Jog NR, Caricchio R. Differential regulation of cell death programs in males and females by Poly (ADP‐Ribose) Polymerase‐1 and 17β estradiol. Cell Death Dis. 2013;4:e758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Caiafa P, Guastafierro T, Zampieri M. Epigenetics: poly(ADP‐ribosyl)ation of PARP‐1 regulates genomic methylation patterns. FASEB J. 2009;23:672–678. [DOI] [PubMed] [Google Scholar]
- 94. Lodhi N, Kossenkov AV, Tulin AV. Bookmarking promoters in mitotic chromatin: poly(ADP‐ribose) polymerase‐1 as an epigenetic mark. Nucleic Acids Res. 2014;42:7028–7038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Lapucci A, Pittelli M, Rapizzi E, Felici R, Moroni F, Chiarugi A. Poly(ADP‐ribose) polymerase‐1 is a nuclear epigenetic regulator of mitochondrial DNA repair and transcription. Mol Pharmacol. 2011;79:932–940. [DOI] [PubMed] [Google Scholar]
- 96. Nalabothula N, Al‐jumaily T, Eteleeb AM, Flight RM, Xiaorong S, Moseley H, et al. Genome‐wide profiling of PARP1 reveals an interplay with gene regulatory regions and DNA methylation. PLoS ONE. 2015;10:e0135410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Caiafa P, Zlatanova J. CCCTC‐binding factor meets poly(ADP‐ribose) polymerase‐1. J Cell Physiol. 2009;219:265–270. [DOI] [PubMed] [Google Scholar]
- 98. Vitale AM, Wolvetang E, Mackay‐Sim A. Induced pluripotent stem cells: a new technology to study human diseases. Int J Biochem Cell Biol. 2011;43:843–846. [DOI] [PubMed] [Google Scholar]
- 99. Doege CA, Inoue K, Yamashita T, et al. Early‐stage epigenetic modification during somatic cell reprogramming by PARP‐1 and Tet2. Nature. 2012;488:652–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. 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] [PubMed] [Google Scholar]
- 101. Huletsky A, de Murcia G, Muller S, et al. The effect of poly(ADP‐ ribosyl)ation on native and H1‐depleted chromatin. A role of poly(ADP‐ribosyl)ation on core nucleosome structure. J Biol Chem. 1989;264:8878–8886. [PubMed] [Google Scholar]
- 102. Wacker DA, Ruhl DD, Balagamwala EH, Hope KM, Zhang T, Kraus WL. The DNA binding and catalytic domains of poly(ADP‐ribose) polymerase 1 cooperate in the regulation of chromatin structure and transcription. Mol Cell Biol. 2007;27:475–7485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Pinnola A, Naumova N, Shah M, Tulin AV. Nucleosomal Core Histones Mediate Dynamic regulation of Poly(ADP‐ribose) Polymerase 1 Protein Binding to Chromatin and Induction of Its enzymatic Activity. J Biol Chem. 2007;282:32511–32519. [DOI] [PubMed] [Google Scholar]
- 104. Krishnakumar R, Gamble MJ, Frizzell KM, Berrocal JG, Kininis M, Kraus WL. Reciprocal binding of PARP‐1 and histone H1 at promoters specifies transcriptional outcomes. Science. 2008;319:819–821. [DOI] [PubMed] [Google Scholar]
- 105. Frizzell KM, Gamble MJ, Berrocal JG, Zhang T, Krishnakumar R, Cen Y. Global analysis of transcriptional regulation by poly (ADP‐ribose) polymerase‐1 and poly (ADP‐ribose) glycohydrolase in MCF‐7 human breast cancer cells. J Biol Chem. 2009;284:33926–33938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Berger SL. Thecomplex language of chromatin regulation during transcription. Nature. 2007;447:407–412. [DOI] [PubMed] [Google Scholar]
- 107. Kotova E, Lodhi N, Jarnik M, Pinnola AD, Ji Y, Tulin AV. Drosophila histone H2A variant (H2Av) controls poly (ADP‐ribose) polymerase1 (PARP1) activation in chromatin. Proc Natl Acad Sci. 2011;108:6205–6210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Kraus WL. New functions for an ancient domain. Nat Struct Mol Biol. 2009;16:904–907. [DOI] [PubMed] [Google Scholar]
- 109. Chen H, Ruiz PD, Novikov L, Casill AD, Park JW, Gamble MJ. MacroH2A1.1 and PARP‐1 cooperate to regulate transcription by promoting CBP‐mediated H2B acetylation. Nat Struct Mol Biol. 2014;21:981–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Krishnakumar R, Kraus WL. PARP‐1 regulates chromatin structure and transcription through a KDM5B‐dependent pathway. Mol Cell. 2010;39:736–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Wu T, Wang XJ, Tian W, Jaramillo MC, Lau A, Zhang DD. Poly (ADP‐ribose) polymerase‐1 modulates Nrf2‐dependent transcription. Free Radic Biol Med. 2014;67:69–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Okada H, Inoue T, Kikuta T, et al. Poly(ADP‐ribose) polymerase‐1 enhances transcription of the profibrotic CCN2 gene. J Am Soc Nephrol. 2008;19:933–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Ong CT, Van Bortle K, Ramos E, Corces VG. Poly(ADP‐ribosyl)ation regulates insulator function and intrachromosomal interactions in Drosophila . Cell. 2013;155:148–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Zhao H, Sifakis EG, Sumida N, et al. PARP1‐and CTCF‐mediated interactions between active and repressed chromatin at the lamina promote oscillating transcription. Mol Cell. 2015;59:984–997. [DOI] [PubMed] [Google Scholar]
- 115. O'Donnell A, Yang SH, Sharrocks AD. PARP1 orchestrates variant histone exchange in signal‐mediated transcriptional activation. EMBO J. 2013;14:1084–1091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Agarwal A, Mahfouz RZ, Sharma RK, Sarkar O, Mangrola D, Mathur PP. Potential biological role of poly(ADP‐ribose) polymerase (PARP) in male gametes. Reprod Biol Endocrinol. 2009;7:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Di Meglio S, Denegri M, Vallefuoco S, Tramontano F, Scovassi AI, Quesada P. Poly(ADPR) polymerase‐1 and poly(ADPR) glycohydrolase level and distribution in differentiating rat germinal cells. Mol Cell Biochem. 2003;248:85–91. [DOI] [PubMed] [Google Scholar]
- 118. Quesada P, Atorino L, Cardone A, Ciarcia G, Farina B. Poly(ADP‐ribosyl)ation system in rat germinal cells at different stages of differentiation. Exp Cell Res. 1996;226:183–190. [DOI] [PubMed] [Google Scholar]
- 119. Schreiber V, Ame JC, Dolle P, Schultz I, Rinaldi B, Fraulob V. Poly(ADP‐ribose) polymerase‐2 (PARP‐2) is required for efficient base excision DNA repair in association with PARP‐1 and XRCC1. J Biol Chem. 2002;277:23028–23036. [DOI] [PubMed] [Google Scholar]
- 120. Meyer‐Ficca ML, Scherthan H, Burkle A, Meyer RG. Poly(ADP‐ribosyl)ation during chromatin remodeling steps in rat spermiogenesis. Chromosoma. 2005;114:67–74. [DOI] [PubMed] [Google Scholar]
- 121. Marcon L, Boissonneault G. Transient DNA strand breaks during mouse and human spermiogenesis new insights in stage specificity and link to chromatin remodeling. Biol Reprod. 2004;70:910–918. [DOI] [PubMed] [Google Scholar]
- 122. Rouleau M, Saxena V, Rodrigue A, et al. A key role for poly(ADP‐ribose) polymerase 3 in ectodermal specification and neural crest development. PLoS ONE. 2011;6:e15834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Rajawat J, Mir H, Begum R. Differential role of poly(ADP‐ribose) polymerase (PARP) in D. discoideum . BMC Dev Biol. 2011;11:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Jubin T, Kadam A, Saran S, Begum R. Poly (ADP‐ribose) polymerase1 regulates growth and multicellularity in D. discoideum. Differentiation 2016;pii:S0301‐4681(15)30081‐5. doi: 10.1016/j.diff.2016.03.002. [DOI] [PubMed] [Google Scholar]
- 125. Couto CA, Wang HY, Green JC, et al. PARP regulates nonhomologous end joining through retention of Ku at double‐strand breaks. J Cell Biol. 2011;194:367–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Mir H, Alex T, Rajawat J, Kadam A, Begum R. Response of D. discoideum to UV‐C and involvement of poly(ADP‐ribose) polymerase. Cell Prolif. 2015;48:363–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Kothe GO, Kitamura M, Masutani M, Selker EU, Inoue H. PARP is involved in replicative aging in Neurospora crassa. Fungal Genet Biol. 2010;47:297–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Müller‐Ohldach M, Brust D, Hamann A, Osiewacz HD. Overexpression of PaParp encoding the poly(ADP‐ribose) polymerase of Podospora anserina affects organismal aging. Mech Ageing Dev. 2011;132:33–42. [DOI] [PubMed] [Google Scholar]
- 129. Semighini CP, Savoldi M, Goldman GH, Harris SD. Functional characterization of the putative Aspergillus nidulans poly(ADP‐ribose) polymerase homolog PrpA. Genetics. 2006;173:87–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. De Block M, Verduyn C, De Brouwer D, Cornelissen M. Poly(ADP‐ribose) polymerase in plants affects energy homeostasis, cell death and stress tolerance. Plant J. 2005;41:95–106. [DOI] [PubMed] [Google Scholar]
- 131. Vanderauwera S, De Block M, Van de Steene N, van de Cotte B, Metzlaff M, Van Breusegem F. Silencing of poly(ADP‐ribose) polymerase in plants alters abiotic stress signal transduction. Proc Natl Acad Sci USA. 2007;104:15150–15155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Fletcher JC, Burtis KC, Hogness DS, Thummel CS. The Drosophila E74 gene is required for metamorphosis and plays a role in the polytene chromosome puffing response to ecdysone. Development. 1995;121:1455–1465. [DOI] [PubMed] [Google Scholar]
- 133. De Murcia JM, Ricoul M, Tartier L, et al. Functional interaction between PARP‐1 and PARP‐2 in chromosome stability and embryonic development in mouse. EMBO J. 2003;22:2255–2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Hamazaki N, Uesaka M, Nakashima K, Agata K, Imamura T. Gene activation‐associated long noncoding RNAs function in mouse preimplantation development. Development. 2015;142:910–920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Bai P, Canto C, Brunyanszki A, et al. PARP 1 regulates SIRT1 expression and whole body energy expenditure. Cell Metab. 2011a;13:450–460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Devalaraja‐Narashimha K, Padanilam BJ. PARP1 deficiency exacerbates diet‐induced obesity in mice. J Endocrinol. 2010;205:243–252. [DOI] [PubMed] [Google Scholar]
- 137. Wang ZQ, Auer B, Stingl L, et al. Mice lacking ADPRT and poly(ADP‐ribosyl)ation develop normally but are susceptible to skin disease. Genes Dev. 1995;9:509–520. [DOI] [PubMed] [Google Scholar]
- 138. Bai P, Canto C, Oudart H, et al. PARP‐1 inhibition increases mitochondrial metabolism through SIRT1 activation. Cell Metab. 2011b;13:461–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Houtkooper RH, Williams RW, Auwerx J. Metabolic networks of longevity. Cell. 2010;142:9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Ying W, Chen Y, Alano CC, Swanson RA. Tricarboxylic acid cycle substrates prevent PARP‐mediated death of neurons and astrocytes. J Cereb Blood Flow Metab. 2002;22:774–779. [DOI] [PubMed] [Google Scholar]
- 141. Andrabi SA, Umanah GKE, Chang C, et al. Poly(ADP‐ribose) polymerase‐dependent energy depletion occurs through inhibition of glycolysis. Proc Natl Acad Sci USA. 2014;111:10209–10214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Bai P, Nagy L, Fodor T, Liaudet L, Pacher P. Poly (ADP‐ribose) polymerases as modulators of mitochondrial activity. Trends Endocrinol Metab. 2015;26:75–83. [DOI] [PubMed] [Google Scholar]
- 143. Asher G, Reinke H, Altmeyer M, Gutierrez‐Arcelus M, Hottiger MO, Schibler U. Poly(ADP‐ribose) polymerase 1 participates in the phase entrainment of circadian clocks to feeding. Cell. 2010;142:943–953. [DOI] [PubMed] [Google Scholar]
- 144. Erener S, Mirsaidi A, Hesse M, et al. ARTD1 deletion causes increased hepatic lipid accumulation in mice fed a high‐fat diet and impairs adipocyte function and differentiation. FASEB J. 2012b;26:2631–2638. [DOI] [PubMed] [Google Scholar]
- 145. Erener S, Hesse M, Kostadinova R, Hottiger MO. Poly(ADP‐ribose)polymerase‐1 (PARP‐1) controls adipogenic gene expression and adipocyte function. Mol Endocrinol. 2012a;26:79–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Hennessy BT, Timms KM, Carey MS, Gutin A, Meyer LA, Flake DD 2nd. Somatic mutations in BRCA1 and BRCA2 could expand the number of patients that benefit from poly(ADP‐ribose) polymerase inhibitors in ovarian cancer. J Clin Oncol. 2010;28:3570–3576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. El‐Khamisy SF, Masutani M, Suzuki H, Caldecott KW. A requirement for PARP‐1 for the assembly or stability of XRCC1 nuclear foci at sites of oxidative DNA damage. Nucleic Acids Res. 2003;31:5526–5533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Houtgraaf JH, Versmissen J, van der Giessen WJ. A concise review of DNA damage checkpoints and repair in mammalian cells. Cardiovasc Revasc Med. 2006;7:165–172. [DOI] [PubMed] [Google Scholar]
- 149. Haince JF, McDonald D, Rodrigue A, et al. PARP‐1 dependent kinetics of recruitment of MRE11 and NBS1 proteins to multiple DNA damage sites. J Biol Chem. 2008;283:1197–1208. [DOI] [PubMed] [Google Scholar]
- 150. Tutt A, Ashworth A. The relationship between the roles of BRCA genes in DNA repair and cancer predisposition. Trends Mol Med. 2002;8:571–576. [DOI] [PubMed] [Google Scholar]
- 151. Arun B, Akar U, Gutierrez‐Barrera AM, Hortobagyi GN, Ozpolat B. The PARP inhibitor AZD2281 (Olaparib) induces autophagy/mitophagy in BRCA1 and BRCA2 mutant breast cancer cells. Int J Oncol. 2015;47:262–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Fong PC, Yap TA, Boss DS, et al. Poly(ADP‐ribose) polymerase inhibition: frequent durable responses in BRCA carrier ovarian cancer correlating with platinum‐free interval. J Clin Oncol. 2010;28:2512–2519. [DOI] [PubMed] [Google Scholar]
- 153. Do K, Chen AP. Molecular Pathways: targeting PARP in cancer treatment. Clin Cancer Res. 2013;19:977–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Gottschalk AJ, Timinszky G, Kong SE, et al. Poly(ADP‐ribosyl)ation directs recruitment and activation of an ATP‐dependent chromatin remodeler. Proc Natl Acad Sci USA. 2009;106:13770–13774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Zaremba T, Ketzer P, Cole M, Coulthard S, Plummer ER, Curtin NJ. Poly(ADP‐ribose) polymerase‐1 polymorphisms, expression and activity in selected human tumour cell lines. Br J Cancer. 2009;101:256–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Espinoza LA. The Role of PARP Activation in Prostate Cancer. Advances in Prostate Cancer, Dr. Gerhard Hamilton (Ed.). Croatia: InTech, 2013. [Google Scholar]
- 157. Mendoza‐Alvarez H, Alvarez‐Gonzalez R. Regulation of p53 sequence‐specific DNA‐binding by covalent poly(ADP‐ribosyl)ation. J Biol Chem. 2001;276:36425–36430. [DOI] [PubMed] [Google Scholar]
- 158. Idogawa M, Masutani M, Shitashige M, et al. Ku70 and poly(ADP‐ribose) polymerase‐1 competitively regulate beta‐catenin and T‐cell factor‐4‐mediated gene transactivation: possible linkage of DNA damage recognition and Wnt signaling. Cancer Res. 2007;67:911–918. [DOI] [PubMed] [Google Scholar]
- 159. Idogawa M, Yamada T, Honda K, Sato S, Imai K, Hirohashi S. Poly(ADP‐ribose) polymerase‐1 is a component of the oncogenic T‐cell factor‐4/beta‐catenin complex. Gastroenterology. 2005;128:1919–1936. [DOI] [PubMed] [Google Scholar]
- 160. Schiewer MJ, Goodwin JF, Han S, Brenner JC, Augello MA, Dean JL. Dual Roles of PARP‐1 Promote Cancer Growth and Progression. Cancer Discov. 2012;2:1134–1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Martire S, Mosca L, d'Erme M. PARP‐1 involvement in neurodegeneration: A focus on Alzheimer's and Parkinson's diseases. Mech Ageing Dev. 2015;146–148:53–64. [DOI] [PubMed] [Google Scholar]
- 162. Moroni F, Cozzi A, Chiarugi A, et al. Long‐lasting neuroprotection and neurological improvement in stroke models with new, potent and brain permeable inhibitors of poly(ADP‐ribose) polymerase. British J Pharmacol. 2012;165:1487–1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Ba X, Gupta S, Davidson M, Garg NJ. Trypanosoma cruzi induces the reactive oxygen species‐PARP‐1‐RelA pathway for up‐regulation of cytokine expression in cardiomyocytes. J Biol Chem. 2010;285:11596–11606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Beneke S. Poly(ADP‐ribose) polymerase activity in different pathologies: the link to inflammation and infarction. Exp Gerontol. 2008;43:605–614. [DOI] [PubMed] [Google Scholar]
- 165. Zhang P, Maruyama T, Konkel JE, et al. PARP‐1 controls immunosuppressive function of regulatory T cells by destabilizing Foxp3. PLoS ONE. 2013;8:e71590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Nasta F, Laudisi F, Sambucci M, Rosado MM, Pioli C. Increased Foxp3+regulatory T cells in poly(ADP‐ribose) polymerase‐1 deficiency. J Immunol. 2010;184:3470–3477. [DOI] [PubMed] [Google Scholar]
- 167. Kaye SB, Lubinski J, Matulonis U, et al. Phase II, open‐label, randomized, multicenter study comparing the efficacy and safety of olaparib, a poly (ADP‐ribose) polymerase inhibitor, and pegylated liposomal doxorubicin in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer. J Clin Oncol. 2011;30:372–379. [DOI] [PubMed] [Google Scholar]
- 168. Mateo J, Friedlander M, Sessa C, et al. Administration of continuous/intermittent olaparib in ovarian cancer patients with a germline BRCA1/2 mutation to determine an optimal dosing schedule for the tablet formulation. Eur J Cancer. 2013;49:S161. [Google Scholar]
- 169. Gelmon KA, Tischkowitz M, Mackay H, et al. Olaparib in patients with recurrent high‐grade serous or poorly differentiated ovarian carcinoma or triple‐negative breast cancer: a phase 2, multicentre, open‐label, non‐randomised study. Lancet Oncol. 2011;12:852–861. [DOI] [PubMed] [Google Scholar]
- 170. Ledermann J, Harter P, Gourley C, et al. Olaparib maintenance therapy in platinum‐sensitive relapsed ovarian cancer. N Engl J Med. 2012;366:1382–1392. [DOI] [PubMed] [Google Scholar]
- 171. Kaufman B, Shapira‐Frommer R, Schmutzler RK, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol. 2015;33:244–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Tutt A, Robson M, Garber JE, et al. Oral poly (ADP‐ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and advanced breast cancer: a proof‐of‐concept trial. Lancet. 2010;376:235–244. [DOI] [PubMed] [Google Scholar]
- 173. Liu JF, Tolaney SM, Birrer M, et al. A Phase 1 trial of the poly(ADP‐ribose) polymerase inhibitor olaparib (AZD2281) in combination with the anti‐angiogenic cediranib (AZD2171) in recurrent epithelial ovarian or triple‐negative breast cancer. Eur J Cancer. 2013;49:2972–2978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Liu JF, Barry WT, Birrer M, et al. Combination cediranib and olaparib versus olaparib alone for women with recurrent platinum‐sensitive ovarian cancer: a randomised phase 2 study. Lancet Oncol. 2014;15:1207–1214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Oza AM, Cibula D, Benzaquen AO, et al. Olaparib combined with chemotherapy for recurrent platinum‐sensitive ovarian cancer: a randomised phase 2 trial. Lancet Oncol. 2015;16:87–97. [DOI] [PubMed] [Google Scholar]
- 176. Rivkin SE, Iriarte D, Sloan H, Wiseman C, Moon J. Phase Ib/II with expansion of patients at the MTD study of olaparib plus weekly (metronomic) carboplatin and paclitaxel in relapsed ovarian cancer patients. ASCO Annu Meet Proc 2014;15(Suppl):5527. [Google Scholar]
- 177. Choy E, Butrynski JE, Harmon DC, et al. Phase II study of olaparib in patients with refractory Ewing sarcoma following failure of standard chemotherapy. BMC Cancer. 2014;14:813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Waxweiler TV, Bowles D, Reddy K, et al. Safety and feasibility update of olaparib, an orally bioavailable PARP inhibitor, with concurrent cetuximab and radiation therapy in heavy smokers with stage III‐IVB squamous cell carcinoma of the head/neck: a phase 1 trial. Int J Radiation Oncol Biol Phys. 2014;90:S559. [Google Scholar]
- 179. Chalmers AJ, Jackson A, Swaisland H, et al. Results of stage 1 of the oparatic trial: a phase I study of olaparib in combination with temozolomide in patients with relapsed glioblastoma. J Clin Oncol. 2014;32:5S. [Google Scholar]
- 180. Samol J, Ranson M, Scott E, et al. Safety and tolerability of the poly(ADP‐ribose) polymerase (PARP) inhibitor, olaparib (AZD2281) in combination with topotecan for the treatment of patients with advanced solid tumors: a phase I study. Invest New Drugs. 2012;30:1493–1500. [DOI] [PubMed] [Google Scholar]
- 181. Coleman RL, Sill MW, Bell‐McGuinn K, et al. A phase II evaluation of the potent, highly selective PARP inhibitor veliparib in the treatment of persistent or recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer in patients who carry a germline BRCA1 or BRCA2 mutation‐An NRG Oncology/Gynecologic Oncology Group study. Gynecol Oncol. 2015;137:386–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Pahuja S, Appleman LJ, Belani CP, et al. Preliminary activity of veliparib (V) in BRCA2‐mutated metastatic castration‐resistant prostate cancer (mCRPC). J Clin Oncol. 2015;33. [Google Scholar]
- 183. LoRusso PM, Tolaney SM, Wong S, et al. Combination of the PARP inhibitor veliparib (ABT888) with irinotecan in patients with triple negative breast cancer: Preliminary activity and signature of response. Cancer Res. 2015;75:CT325. [Google Scholar]
- 184. Owonikoko TK, Dahlberg SE, Khan SA, et al. A phase 1 safety study of veliparib combined with cisplatin and etoposide in extensive stage small cell lung cancer: A trial of the ECOG–ACRIN Cancer Research Group (E2511). Lung Cancer. 2015;89:66–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. McKee MD, Bondarenko I, Guclu SZ, et al. Veliparib (ABT‐888) or placebo combined with carboplatin and paclitaxel in patients with previously untreated advanced/metastatic squamous (Sq) non‐small cell lung cancer (NSCLC): a randomized phase 3 trial. J Clin Oncol. 2015;33. [Google Scholar]
- 186. Kummar S, Ji J, Morgan R, et al. A Phase I Study of Veliparib in Combination with Metronomic Cyclophosphamide in Adults with Refractory Solid Tumors and Lymphomas. Clin Cancer Res. 2012;18:1726–1734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. O'Reilly EM, Lowery MA, Segal MF, et al. Phase IB trial of cisplatin (C), gemcitabine (G), and veliparib (V) in patients with known or potential BRCA or PALB2‐mutated pancreas adenocarcinoma (PC). ASCO Annual Meeting Proceedings 2014;32(15 suppl):4023. [Google Scholar]
- 188. Hussain M, Carducci MA, Slovin S, et al. Targeting DNA repair with combination veliparib (ABT‐888) and temozolomide in patients with metastatic castration‐resistant prostate cancer. Invest New Drugs. 2014;32:904–912. [DOI] [PMC free article] [PubMed] [Google Scholar]
