You are on page 1of 12

Review articles

The PARP superfamily


Jean-Christophe Amé, Catherine Spenlehauer, and Gilbert de Murcia*

Summary and Mandel correctly anticipated the major property of an


Poly(ADP-ribosyl)ation is an immediate DNA-damage- enzymatic activity involved in DNA-dependent NADþ con-
dependent post-translational modification of histones
and other nuclear proteins that contributes to the survival sumption. This seminal contribution triggered the science of
of injured proliferating cells. Poly(ADP-ribose) poly- poly(ADP-ribose) (PAR) that today covers a large area of
merases (PARPs) now constitute a large family of 18 biology dealing with genomic stability and energy metabolism.
proteins, encoded by different genes and displaying a Poly(ADP-ribosyl)ation of nuclear proteins is a post-transla-
conserved catalytic domain in which PARP-1 (113 kDa), tional modification induced by DNA strand breaks that
the founding member, and PARP-2 (62 kDa) are so far the
sole enzymes whose catalytic activity has been shown to establishes a molecular link between DNA damage and
be immediately stimulated by DNA strand breaks. A large chromatin modification. A large body of evidence demon-
repertoire of sequences encoding novel PARPs now strates that this activity efficiently contributes to a detection–
extends considerably the field of poly(ADP-ribosyl)ation signaling pathway leading ultimately to the resolution of
reactions to various aspects of the cell biology including strand-break interruptions. In certain pathophysiological con-
cell proliferation and cell death. Some of these new
ditions, this protecting function is strongly activated leading to
members interact with each other, share common part-
ners and common subcellular localizations suggesting cell death, tissue damage and organ failure.
possible fine tuning in the regulation of this post- From genome sequencing and various molecular and
translational modification of proteins. This review sum- genetic approaches, it became evident that more than one
marizes our present knowledge of this emerging super- enzyme was involved in the conversion of NADþ to ADP-
family, which might ultimately improve pharmacological
strategies to enhance both antitumor efficacy and ribose polymers. A superfamily of eighteen proteins containing
the treatment of a number of inflammatory and neurode- PARP domains has recently emerged. In this review, we have
generative disorders. A provisional nomenclature is summarized our present knowledge of this new family of
proposed. BioEssays 26:882–893, 2004. proteins, highlighting some of the yet unexplored functions of
ß 2004 Wiley Periodicals, Inc. PARP-1, its founding member.

Introduction The PARP superfamily: seven isolated cDNAs


Forty years ago, in their initial paper entitled ‘‘Nicotinamide The characterization of the human PARP family of proteins
mononucleotide activation of a new DNA-dependent poly- was based on an exhaustive search of the non-redundant
adenylic acid synthesizing nuclear enzyme’’ Chambon, Weil protein database (NCBI) using the human PARP-1 catalytic
domain (GenBank XP_037275 residues 796–1014). Eighteen
Unité 9003 du CNRS, École Supérieure de Biotechnologie de different putative PARP homologues were found (Fig. 1) that
Strasbourg, Illkirch, France. could potentially carry poly(ADP-ribose) polymerase activity.
Both authors contributed equally to this work.
Although, we are far from understanding the structure and
Funding agency: Centre National de la Recherche Scientifique,
Association pour la Recherche Contre le Cancer, Électricité de
function of all members of the PARP family, biochemical and
France, Ligue Nationale Contre le Cancer and Commissariat à enzymatic properties have been studied for at least seven of
l’Énergie Atomique. them to various degrees, giving a provisional picture of this
*Correspondence to: Dr. Gilbert de Murcia, Unité 9003 du CNRS, rapidly growing area of post-translational modification of
École Supérieure de Biotechnologie de Strasbourg, Boulevard
proteins.
Sébastien Brant, BP 10413, F-67412 Illkirch Cedex, France.
E-mail: demurcia@esbs.u-strasbg.fr
PARP-1, the founding family member, has been the most-
DOI 10.1002/bies.20085 extensively studied. In response to DNA damage, it uses
Published online in Wiley InterScience (www.interscience.wiley.com). NADþ to synthesize a linear or multibranched polymer of ADP-
ribose on various nuclear protein acceptors usually associated
with chromatin, or, itself in an automodification reaction. Both
automodification and heteromodification of acceptors con-
Abbreviations: PARP, poly(ADP-ribose) polymerase; XRCC1, x-ray
tribute positively to the survival of injured proliferating cells
cross complementing factor 1; BRCT, BRCA1 c-terminus; GFP, green
fluorescent protein; 3-AB, 3-aminobenzamide;; DAPI, 40 ,6-diamidino-
(see for review(1,2)). The structure–function relationship of
2-phenylindol; SSB, single strand breaks; SSBR, single strand breaks PARP-1 is now well understood, four domains out of six
repair; DSB, double strand breaks. have been assigned a particular function: (1) domain A, the
DNA-binding domain (Fig. 1), containing two zinc finger

882 BioEssays 26.8 BioEssays 26:882–893, ß 2004 Wiley Periodicals, Inc.


Review articles

Figure 1. The PARP superfamily. Schematic domain architecture of the eighteen members of the poly(ADP-ribose) polymerase (PARP)
superfamily. The proteins are drawn to scale and low-complexity segments in certain proteins have been omitted. Each protein has been
locally searched against the Pfam database version 7(35) using the program hmmpfam of the HMMER2 package. The most significant
domains detected have been indicated with colored boxes and their limits indicated by the position numbers. Here is a brief description of the
most significant one. The BRCT (BRCA1 C Terminus) domain is found predominantly in proteins involved in cell-cycle-checkpoint functions
responsive to DNA damage. The WWE domain is named after three of its conserved residues and is predicted to mediate specific protein–
protein interactions in ubiquitin and ADP ribose conjugation systems. The UIM motif is called the ubiquitin interaction motif. The SAP (after
SAF-A/B, Acinus and PIAS) motif is a putative DNA -binding domain found in diverse nuclear proteins involved in chromosomal organiza-
tion. The macro-H2A (A1pp) domain is found at the C terminus of the macro-H2A histone protein and is a predicted phosphoesterase
domain that is supposed to function as an ADP-ribose phosphoesterase that might regulate the protein(ADP-ribosyl)ation.(36) The RRM
motif is probably diagnostic of an RNA -binding protein. The dendrogram was based on the alignment of Fig. 2 and was generated with the
JALVIEW program.

structures responsible for PARP-1’s interaction with DNA features with the active site of bacterial (ADP-ribosyl)ating
breaks, which are also found in DNA ligase III and in the 30 DNA toxins (Diphteria toxin or Pertussis toxin) notably the NAD-fold:
phosphoesterase from Arabidopsis thaliana (see for re- b-a-loop-b-a corresponding, in PARPs, to the most conserved
view(3)), (2) domain B containing a bipartite nuclear localiza- region (PARP signature) (Fig. 2a,b). It contains an additional
tion signal (NLS) responsible for the nuclear homing of PARP- a-helical domain (a-hd) that is thought to relay the activation
1 and a caspase-3 cleavage site,(4) (3) domain D, the signal issued on binding to damaged DNA. Nothing is known
automodification domain, containing a BRCT motif constitut- so far concerning the function of domains C and E.
ing the major protein interface with various nuclear partners, The role of PARP-1 in facilitating DNA repair has been
and (4) domain F, the smallest PARP-1 fragment retaining clearly demonstrated by the generation of three independent
catalytic activity (see for review(5)). The crystal structure of the PARP-1-deficient mouse models (see for review(7)). These
chicken PARP-1 catalytic domain(6) shows common structural animals showed hypersensitivity to ionizing radiation and

BioEssays 26.8 883


Review articles

Figure 2. The PARP signature. a: Multiple alignment of the eighteen human PARP sequences. The secondary structure of the domain as
determined from the chicken PARP catalytic domain(6) has been superimposed under the alignment with the following code: blue arrows for
b sheets, red cylinder for a helices and straight lines for loops. The amino acid color code is that of CLUSTALX(74) (blue: >60% hydrophobics
(ACFHILMVWY); Magenta: >50% negative charges (DE); Red: 60% positive charges (KR); Green: >50% polar (STQN); Pink: >85%
cysteines; Orange: >8% glycines; Yellow: >85% prolines; Cyan: >50% Aromatics (FYW)). The sequences are denoted by their common
laboratory name. b: Ribbon representation of the PARP signature.

alkylating agents at the cellular or whole-animal level. PARP-2 whose function is still not clear acts as a chromatin
Unexpectedly, these animals also showed protection against modifier as mentioned above. PARP-2 interacts with PARP-1
various inflammatory processes such as cerebral and cardiac and shares common partners involved in the Single Strand
ischemia and a higher resistance to septic shock (see for Break Repair (SSBR) and Base Excision Repair (BER)
reviews(2,8)). pathways: XRCC1, DNA polymerase-b, and DNA ligase III.
PARP-2 was discovered as a result of the presence of PARP-1 and PARP-2 also interact with proteins involved in the
residual DNA-dependent PARP activity in embryonic fibro- kinetochore structure and in the mitotic spindle checkpoint
blasts derived from PARP-1-deficient mice.(9,10) Its catalytic (see below). However, specific partners of PARP-2 are
domain has the strongest resemblance to that of PARP-1 with beginning to be discovered, such as the telomeric protein
69% similarity. The crystal structure of the murine PARP-2 is TRF2, suggesting a link with telomere integrity control.(14) The
very similar to that of PARP-1 but with the exception of involvement of PARP-2 in the cellular response to DNA
differences in the vicinity of the acceptor site that reflect dif- damage has been investigated by the generation of a PARP-2-
ferences in terms of substrates.(11) Clearly, PARP-2 prefer- deficient mouse model.(15) Following treatment by the alkylat-
entially heteromodifies histone H2B (Huber et al., unpublished ing agent N-nitroso-N-methylurea (MNU), PARP-2-deficient
results) whereas PARP-1 preferentially modifies histone cells displayed a large delay in DNA-strand breaks resealing,
H1(12) in isolated nuclei. PARP-2 is a nuclear protein that similar to that observed in PARP-1-deficient cells,(16) thus
binds to and is activated by DNase-I treated DNA, however, confirming that PARP-2 is an active player in SSBR in spite of
its DNA-binding domain differs from that of PARP-1 and it its reduced capacity to synthesize ADP-ribose polymers.
targets DNA gaps but not nicks (J.C.A., unpublished data). However, PARP-2 deficient mice also display specific pheno-
This domain, containing 75 aa, localizes the protein to the types (see below), indicating that PARP-1 and PARP-2
nucleus and displays some homology with the SAP domain(13) functions are complementary but do not fully overlap.
found in various nuclear proteins, such as AP-endonuclease PARP-3 has been identified as a core component of
and Ku70, involved in chromosomal organization and in DNA the centrosome(17) preferentially located at the daughter
repair. Interestingly, this domain is also found duplicated in centriole throughout the cell cycle. Of all the PARPs, it dis-
plant PARP-2 orthologs (Arabidopsis thaliana #NP_192148 plays the smallest N-terminal domain with only 54 residues
and Zea mays #T03656). responsible for the centrosomal localization. PARP-3 shares

884 BioEssays 26.8


Review articles

with PARP-1 two domains, E and F, which display a 61% ratory: shutting down Tankyrase 1 expression by siRNA blocks
similarity. PARP-3 activity is hardly detectable but can never- the cells during early anaphase because sister telomeres are
theless be inhibited by the PARP inhibitor 3-aminobenzamide. unable to segregate, indicating that the replicated telomeres
PARP-3 interacts with PARP-1 which, in part, also resides in are held together by specific cohesion complexes that are
the centrosome(18) suggesting a link between the DNA- resolved by Tankyrase 1.(26)
damage surveillance network and the mitotic fidelity check- A second tankyrase (PARP-5b or tankyrase-2) encoded by
point (see below). a distinct gene has also been discovered.(27) The sequence of
VPARP (PARP-4), the largest of the family (192.6 kDa), this protein exhibits >85% identity with tankyrase-1 but lacks
was discovered associated with vault particles, a cytoplasmic the HPS motif. Tankyrase 1 and Tankyrase 2 interact with the
ribonucleoprotein complex that associates two other highly same set of proteins (TRF1, IRAP, Glut4, Grb14, TAB 182 and
conserved proteins, major vault protein (MVP) and telomer- NuMa(28–33) and probably mediate overlapping functions in
ase-associated protein (TEP1) and an untranslated vault telomere homeostasis and vesicle trafficking (glucose trans-
RNA (VRNA).(19) The function of these unusual particles is port and insulin signaling). Accordingly, Tankyrase 1 and
unknown, but they may play a role in cellular transport and Tankyrase 2 show multiple subcellular localizations: telome-
could be associated with multidrug resistance in some cell res, nuclear pores, Golgi vesicles and pericentriolar material
lines. The protein has also been localized to nuclear pores and (PCM) during mitosis. Tankyrase 2, however, displays distinct
is associated with the mitotic spindle during mitosis. The properties from those of tankyrase 1. When overexpressed, it
domain structure is quite unusual since domains E and F are induces caspase-independent cell death through the loss of
located at the N-terminal part of the protein. VPARP as well as mitochondrial potential, a process inhibited by the PARP
purified vault particles are capable of catalyzing the poly(ADP- inhibitor 3-aminobenzamide.(27)
ribosyl)ation of MVP. PARP-4 contains a BRCT-motif-contain- TiPARP (PARP-7) was identified by differential display
ing domain at the most N-terminal part of the protein and the as a 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)-induced
type A domain of the von Willebrand factor whose common mRNA.(34) The exact function of TiPARP remains unclear. It
feature appears to be involvement in multiprotein complexes. seems to be involved in T-cell function, and its induction by
It has recently been shown that mVparp, alone or in TCDD contributes to tumor promotion. The in vitro transcribed/
combination with mTep1, is dispensable for normal embryonic translated protein appears to be able to catalyze histone
development, telomerase catalysis, telomere length main- poly(ADP-ribosyl)ation despite the absence of the catalytic
tenance, and vault structure in vivo.(77) Glu residue equivalent to Glu 988 of PARP-1.
Tankyrase 1(TRF1-interacting, ankyrin-related ADP-ri-
bose polymerase), (PARP-5a), was identified as a partner of More PARPs from human
the human telomeric protein TRF1 in a two-hybrid screen- genome sequencing
ing.(20) It contains the smallest domain homologous to PARP-1 To investigate further the possible biological role of the other
that still displays PARP enzymatic activity. The catalytic site members of the PARP family, we looked at their potential
spans only 240 aa compared to 353 aa for PARP-1. The a-hd domain organization by searching the latest Pfam(35) database
domain (Fig. 1), comprising a succession of five N-terminal using the HMMER hidden Markov model software (http://
helices in the PARP-1 crystal structure, is replaced in hmmer.wustl.edu). The surprising feature of these proteins is
Tankyrase by the SAM (Sterile Alpha Motif) domain also the diversity of the new domains that the PARP catalytic
made of a series of five helices arranged in a small globular domain is associated with, suggesting that they may be
domain. This domain acts as a protein-interaction module implicated in many biological functions (Fig. 1). These proteins
through its ability to homo- and hetero-oligomerize with other are distinct from the previously described PARPs in that they
SAM domains including Tankyrase 2.(21) The major part of the combine their catalytic domains with a variety of adapter
protein, spanning 843 aa, is made of Ankyrin repeats orga- domains such as zinc fingers, SAP, Ankyrin, BRCT, Macro
nized in five clusters(22) involved in its interaction with partners. domain and SAM.
In addition, the N-terminal domain contains homopolymeric The most-frequent domain found in five of the new PARPs
runs of His, Pro and Ser (HPS) harboring a MAP kinase (PARP-7, PARP-11 to PARP-14) is the WWE domain occuring
consensus motif (PXSP). The phosphorylation of this motif in classes of proteins associated with ubiquitination.(36) The
upon insulin stimulation enhances Tankyrase 1 activity and link with ubiquitination is also found in the Ubiquitin Interaction
suggests that it is an important insulin signaling target.(23) Motif (UIM) present in PARP-10. Three of the new PARPs
Overexpression of Tankyrase 1 promotes ADP-ribosylation of (PARP-7, PARP-8 and PARP-13) have one zinc finger and
TRF1, leading to its release from telomeres and to telomere PARP-12 has three zinc fingers that differ from the zinc finger
elongation(24) after which Tankyrase is finally ubiquitinated found in PARP-1 and in DNA-ligase III. The zinc fingers of
and degraded by the proteasome.(25) The in vivo function of PARP-7, PARP-12 and PARP-13 are of the C-x8-C-x5-C-x3-H
Tankyrase 1 has been recently clarified by the Smith’s labo- type, which includes zinc finger proteins from eukaryotes

BioEssays 26.8 885


Review articles

involved in cell cycle or growth-phase-related regula- The PARP active site:


tion. PARP-10 and PARP-15 have a motif specific for binding a conserved core in the family members
to RNAs with the RNA Recognition Motif (RRM) found in The sequence alignment relative to the 18 PARPs catalytic site
a variety of RNA-binding proteins, including heterogeneous is presented in Fig. 2a. It reveals major conservation blocks
nuclear ribonucleoproteins (hnRNPs), proteins implicated defining the ‘PARP signature’ that coincide with key secondary
in regulation of alternative splicing, and protein compone- structures constituting the active site. The highest sequence
nts of small nuclear ribonucleoproteins (snRNPs). This and length variability is observed between positions 70 and
motif also appears in certain single-stranded DNA-binding 105 and between positions 168 and 202 of the alignment that
proteins. introduces some major gaps. The flexibility of these regions is
PARP-9 (bal) has recently been discovered in patients with consistent with the crystal structure of the PARP domain(6) as
certain types of diffuse large B-cell lymphomas (DLBCL). It has these regions represent loops of variable length that may act
been described as a new nuclear protein encoded by the BAL as interacting sites to allow the recruitment of acceptor
gene, which is expressed at a significantly higher level in fatal proteins (Fig. 2b). The catalytic residue E988 of PARP-1 in
high-risk DLBCLs and may be implicated in B-cell migra- position 214 of the alignment has been found to be conserved
tion.(37) The overexpression of BAL increases the rate of among PARP-1s from various species, in PARP-2, PARP-3,
migration of B-cell lymphoma transfectants, suggesting that PARP-4 and in Tankyrases (PARP-5a and b). This Glu residue
the risk-related protein may promote the dissemination of high- is also conserved in four of the new members: PARP-5c,
risk DBLCls. This protein contains a duplicated A1pp domain, PARP-11, PARP-12 and PARP-14. In PARP-6 and PARP-8
also called the macro domain, found in a number of otherwise the Glu residue appears at a different position and could be
unrelated proteins. PARP-14 and PARP-15 also contain this replaced by an Asp in PARP-15, but neither the Glu nor the Asp
duplicated domain found at the C terminus of the macro-H2A residues are present in PARP-7, PARP-9, PARP-10, PARP-
histone protein and in the non-structural proteins of several 13 and PARP-16. For these later PARPs, no other consensus
types of ssRNA viruses. It is also found on its own in a family of residue appears to show up with the exception of a Pro residue
proteins from bacteria (P75918), archæbacteria (O59182) and at position 213, which is observed in eleven of them (the three
eukaryotes (Q17432), suggesting that it is involved in an hPARP-5 and PARP-7, PARP-9 to PARP-15). The absence of
important and ubiquitous cellular process. The recent crystal- the Glu catalytic residue may have an impact in limiting the
lization of AF1521, a protein consisting of a stand-alone macro elongation activity to the synthesis of mono-ADP-ribose. The
domain from Archaeoglobus fulgidus shed some light on its majority of the products detected in vivo by immunological
function.(38) Its structure closely resembles the N-terminal staining (using polyclonal antibodies directed against ADP-
DNA-binding domain of Escherichia coli leucine aminopepti- ribose polymers) appear to be shorter polymers.(39) The
dase PepA and shows some similarity to members of the biological importance of these PARPs lacking the Glu catalytic
P-loop family of nucleotide hydrolases, like the phosphoes- residue should therefore not be neglected despite their
terases that act on ADP-ribose derivatives. Though it is possibly reduced enzyme activity.
interesting to find the macro domain associated with potential The dendrogram (Fig. 1) based on the alignment shows the
PARP domains, the sequence variation amongst the macro relationship between the various PARPs. Three groups can be
domain sequences is so high compared to that of the AF1521 distinguished: the PARP-1 group containing PARP-2, PARP-3,
family that it is not clear if they still retain catalytic activity. It is PARP-4, PARP-6, PARP-8 and PARP-16, the Tankyrase group
however possible that, in these proteins, the macro domain (PARP-5a, b, c) and the last group with the members PARP-7,
functions as a nucleic-acid-binding module as is the case with and PARP-9 to PARP-15. Thus, despite its comparatively short
the PepA N-terminal domain. length, the PARP motif contains sufficient variation to allow
These observations suggest that the new domains function phylogenetic discrimination. Remarkably, the catalytic domain
in specific interaction with other proteins and could help in seems also to follow the presence of specific domains such
targeting the different kinds of poly(ADP-ribosyl)ation activ- as the macroH2A and the WWE domains that are distributed
ities of proteins participating in various signaling cascades. within two separate subgroups: PARP-9, PARP-10, PARP-14
An interesting feature of the PARP family is the capacity of and PARP-15 and PARP-7, PARP-11, PARP-12, and PARP-
some of its members to interact with each other, like PARP-1 13, respectively. Table 1 summarizes the main features of the
with PARP-2(16) and PARP-3(17) and Tankyrase 1 with genes and gene products forming the PARP family.
Tankyrase 2.(28,32) This type of combination/association
could involve these complexes in different subcellular com- PARPs and the mitotic apparatus
partments and with different substrates, which they could Mitosis is the most dramatic and potentially dangerous event in
not otherwise target as individuals. This may highlight a new the cell cycle as sister chromatids are irreversibly segregated
organizational order for the PARPs that could diversify greatly in daughter cells. It is therefore not surprising to find several
their biological responses, via combinatory interactions. PARPs associated with centrosomes, telomeres, centromeres

886 BioEssays 26.8


Review articles

Table 1. The table summarizes the main features of the PARP family sequences and gives their corresponding
GenBank identifier
Proposed Expression
nomenclature MW Aa Ip Accession# Chromosome % of PARP-1 % ident. % simil.

hPARP-1 113083 1014 9,3 XM_037275.1 1q41-42 100,0 100 100


hPARP-2 64819 570 8,6 Q9UGN5 14q11.2 14,9 46 69
hPARP-3 60088 533 6,7 XM_052494 3p21 16,6 39 61
hPARP-4 (VPARP) 192672 1724 5,3 AF158255 13q11 38,3 34 54
hPARP-5a (tankyrase1) 142011 1327 7,0 NM_003747.1 8p23.1 6,5 38 52
hPARP-5b (tankyrase2) 126917 1166 7,2 AF309033.1 10q23.03 34 54
hPARP-5c (tankyrase3) 29552 261 8,2 AK023746.1 38 54
hPARP-6 71144 630 8,4 AK022547 15q22.3 34,1 29 51
hPARP-7 (TiPARP) 78926 680 6,4 AL080156.1 3q25.31 24,4 26 50
hPARP-8 55824 501 9,5 XM_018395.2 5q11.2 9,9 25 48
hPARP-9 (Bal) 96282 854 7,9 NM_031458.1 3q13-q21 23,8 27 57
hPARP-10 109327 1020 4,6 NM_032789.1 8q24.3 35,8 28 47
hPARP-11 38738 331 7,8 AF263540 12p13.3 2,4 35 58
hPARP-12 79063 701 8,6 XM_034821.1 7q34 23,9 33 54
hPARP-14 170600 1518 8,4 XM_291055 3q21.1 31,0 30 47
HparP-15 111765 989 8,0 XM_093336 3q21.1 92,2 22 41
HparP-16 36382 322 9,4 XM_113792 15q22.2 13,6 21 32

For each sequence, the Molecular Weight (MW), the Amino acid length (Aa), and the isoelectric point (Ip) of the protein are indicated. The chomosomal
localization of the corresponding gene is based on build 33 of the NCBI human genetic map. The cDNA expression of the various PARPs is the % relative to that
of PARP-1 cDNA expression, it was established by searching the NCBI est_human database with BLAST using the corresponding cDNA. The amino acid
conservation (% of identity and % of similarity) is given only for the PARP catalytic domain relative to that of PARP-1.

and microtubules (Fig. 3). Recent studies on syncytial Parvinen, G. de M. and P. Sassone-Corsi, unpublished data)
Drosophila embryos demonstrated that centrosome inactiva- are clearly involved in the organization of pericentric chromatin
tion is part of the mitotic response to DNA lesions that blocks and in the mechanics of cell division. In response to DNA
chromosome segregation and prevents proliferation of cells damage during mitosis, poly(ADP-ribosyl)ation of Aurora B by
with genomic instability.(40) This response appears to require PARP-1 and/or PARP-2 has been shown to negatively
the localization of the checkpoint kinase Chk2 to the centro- regulate its kinase activity on Ser10 of Histone H3, thus halting
some. It remains to be determined whether higher eukaryotes metaphase progression (L. Monaco, R. Loury, J. Ménissier-de
show a comparable centrosomal response to impaired DNA Murcia, M. Parvinen, G. de M. and P. Sassone-Corsi,
integrity and if PARP-1 and/or PARP-3 play a role in this unpublished data). In addition, the presence in the midbody
process. of PARP-1 and PAR following DNA damage (C.S. and G. de M.
Remarkably, a number of recently identified partners of unpublished results) suggest a possible function in a detec-
PARP-1 and PARP-2 such as CENP-A, CENP-B, Bub 3(41–43) tion–signaling pathway aimed at monitoring the eventual
and Aurora B (L. Monaco, R. Loury, J. Ménissier-de Murcia, M. presence of broken DNA that originates from tension forces

Figure 3. The PARP superfamily and the cell


division. Subcellular localization of some of the
PARP family members to the mitotic apparatus
(see text for details).

BioEssays 26.8 887


Review articles

between two daughter cells experiencing unbalanced chro- Ser139(45) are two epigenetic marks induced by DNA strand
mosome segregation. Therefore, PARP-1 displays most of the breaks that contribute to the histone code in damaged
characteristic behavior of a chromosomal passenger that chromatin. DNA-damage-induced poly(ADP-ribosyl)ation of
associates with kinetochores of cells prepared for nuclear H1 and H2B contributes to the relaxation of the 30 nm
division and later on localizes to the bundled filaments forming chromatin fibre in vitro (Fig. 4) and we have proposed a long
the central spindle and finally the contractile ring. The various time ago that this immediate conformational change of the
localizations of PARP-1 during the metaphase–anaphase nucleosomal superstructure may trigger the access of repair
transition as well as the anaphase–telophase transition enzymes to the lesion in vivo.(12) Non-covalent interactions
strongly suggest a possible role in maintaining the genome between histone tails and PAR also take place, further
stability and segregation fidelity during cell division by con- decreasing nucleosomal stability.(46)
troling the cell cycle progression at various stages in response The role of PARP-1 during the repair of single-strand
to DNA damage. breaks in mammalian cells is now better understood, not only
PARP-2 also localizes to centromeres during prometa- in vitro where PARP-1 activity facilitates the whole process,
phase and shares with PARP-1 the same partners in this but also in living cells where PAR synthesis at sites of DNA
compartment: CENP-A, CENP-B and Bub3.(43) Interestingly, lesions plays an essential role in the recruitment kinetics of a
the disruption of the mouse parp-2 gene lead to sensitization to key player: XRCC1.(47,48) This remarkable property observed
ionizing radiation and monoalkylating agents(15) associated by Okano et al. is illustrated in Fig. 4, but with a different
with preferential centromeric breaks, a prominent G2/M experimental setting. A robust PAR synthesis is immediately
accumulation, acquisition of 8N DNA content and a significant detected in proliferating cells irradiated with a laser microbeam
level of anaphase bridges. The elevated rate of chromosome at 337 nm in the presence of Hoechst. The absence of dynamic
mis-segregation is reminiscent of kinetochore defects that recruitment of XRCC1 in PARP-1-deficient cells could thus
take place in PARP-2-deficient mice irradiated with low doses explain the important delay in strand-break rejoining that, in
of g-rays. In summary, these data suggest that the presence of turn, causes a severe DNA repair defect.(49)
several PARPs at kinetochores, makes these key regions of PARP-1 activity also occurs during the repair of damaged
our chromosome-sensitive integrators of DNA strand breaks, bases by BER. Activation of polymer synthesis likely follows
capable of instantly interrupting the process of microtubule the incision step of the sugar-phosphate backbone by DNA
capture to prevent mitotic errors. glycosylase-AP lyases or APE-1. We have shown that the
Double mutant mice (parp-1/parp-2/) are not viable polymerization step of the Long Patch Repair (LPR) pathway
and die at the onset of gastrulation demonstrating that DNA- was mainly affected in PARP-1-deficient cells.(50) Moreover,
dependent poly(ADP-ribosyl)ation is essential during early PARP-1 is associated with DNA pol-b(50,51) and efficiently
embryogenesis. Surprisingly, specific female embryonic leth- binds to the repair intermediates containing a flap50 -abasic site
ality is observed in parp-1þ/parp-2/ (but not in parp-1/ that are formed before sub-pathway choice, leading to either
parp-2þ/ mice) accompanied by a specific instability of Short Patch Repair (SPR) or LPR.(51,52) The ligation step is
the X chromosome.(15) A defective segregation of the also affected by the presence of PARP-1 since it was demo-
X chromosome, already known as a lagging chromosome, nstrated that a direct physical interaction between PARP-1
can be evoked. The already known lagging character of and DNA ligase III occurs via the region of amino acids
the inactivated X chromosome(44) could be associated with the immediately adjacent to the N-terminal zinc finger of DNA
fact that, as a silenced region of the genome, it may be less ligase III.(53) It was further demonstrated that DNA ligase III
frequently repaired than actively transcribed portions and also binds directly to PAR and to poly(ADP-ribosyl)ated
therefore needs to be repaired during DNA replication to be PARP-1, leading to an increase in DNA joining. This provides
faithfully transmitted. Depending on the extent of DNA a mechanism for the recruitment of the DNA ligase III–XRCC1
damage, the inactivated X chromosome might not be captured complex at DNA breaks. Therefore the presence of PARP-1
in time by the microtubules, this delay is most probably and/or PAR throughout the BER/SSBR process, in interaction
accentuated in the parp-2/ context, thus increasing its with XRCC1 and DNA Ligase III, which are not found in lower
instability. eukaryotes, suggests a unique role in maintaining genome
stability in complex organisms.
The dual role of poly(ADP-ribose) It is not known whether the phosphorylation of histone
synthesis in response to DNA strand breaks: H2AX on Ser139 also contributes to chromatin accessibility
chromatin loosening and XRCC1 recruitment after damage. It seems rather to induce the recruitment of
Changes in chromatin structure resulting from DNA breaks are double-strand-break repair factors and checkpoint control
probably the earliest events in the cellular response to DNA proteins at damaged foci providing a means of ordering the
damage. Poly(ADP-ribosyl)ation of histones and nuclear molecular events ensuing from DSB detection.(54,55) In
proteins as well as phosphorylation of histone H2AX on fact, the epigenetic mark put to histone H2AX in response to

888 BioEssays 26.8


Review articles

Figure 4. The multiple functions of PAR in the cellular response to DNA damage. In response to DNA damage, DNA-damage-dependent
PARPs detect(75) and signal interruptions in the double helix. DNA-break-induced poly(ADP-ribosyl)ation of histone H1 and PARPs leading,
in a few seconds, to the relaxation of chromatin structure(12,76) and simultaneously to the recruitment of XRCC1(47) and SSBR enzymes to
the damage site, thus increasing the repair kinetics. The native chromatin structure (30 nm fibre) is finally restored after degradation
of PAR by PARG. Under certain physiological conditions, the persistence of free polymers (PAR) may have a stress-signaling function
(see also Fig. 5).

DNA-strand break can occur independently of PAR synthesis heat-shock genes become decondensed, swell to many times
or of the PARP-1 and PARP-2 status.(56) Thus, co-localization their original diameter, and accumulate many new mRNA
but no connection apparently occurs between these two post- transcripts. The same puffs occur at chromosomal sites of
translational modifications acting on chromatin structure in hormone-responsive genes in response to increased pro-
response to DNA breakage. It is worth recalling that the duction of steroid hormone in larvae approaching pupation.
simultaneous loss of both modifications in the double mutation Tulin and Spradling(58) showed that PARP-1 is crucial for puff
(parp-1/; atm/)(57) leads to early embryonic lethality formation in Drosophila polytene chromosomes. PARP-1 is
suggesting the absolute necessity of maintaining at least one widely distributed on these chromosomes and is normally
of these two important pathways, especially during early inactive. However, after a heat-shock stimulus, the protein
embryonic development. accumulates rapidly at heat-shock loci where an intense PAR
synthesis activity occurs. A similar PARP accumulation and
The case of D. melanogaster activation is noted in ecdysone-induced puffs in larvae before
Recent studies have shown that PARP-1 and/or its enzymatic pupation. Another target for PARP-1 is the nucleolus, where
activity are directly involved in transcription regulation in rRNA genes are the sites of the major transcriptional activity of
Drosophila where it is possible to visualize the activity of genes the cells even though they are surrounded by condensed
at specific loci on polytene chromosomes.(58) In addition, heterochromatin that usually inhibits transcription. Transcrip-
Drosophila is a suitable model to study PARP biology because tion and puffs formation is largely reduced by PARP-1
it contains just two PARP genes, a PARP-1 like gene and mutations or by the PARP inhibitor 3-aminobenzamide. The
tankyrase. In the fruit fly, the transcriptional response is rapid, study reveals a strong correlation between PAR accumulation
producing many mRNA transcripts, fading shortly after the and puffing. In addition, the authors found that PARP mutant
inducing signal disappears. This rapid transcriptional re- Drosophila are extremely sensitive to bacterial infections
sponse is clearly visible under the microscope as ‘‘puffs’’ in due to a defective activation of genes involved in the control
the giant polytene chromosomes of the salivary glands. Within of the immune response, similar to NF-kB homologues
minutes of a heat-shock stimulus, the chromosomal sites of rejoining the phenotypes observed for the PARP-1 knockout

BioEssays 26.8 889


Review articles

mice.(59) These observations argue that PARP’s role in NF-kB- normally confined to the mitochondrial intermembrane space
dependent immune response gene expression has been in healthy cells. On induction of cell death by several
conserved during evolution. PARP-1 may act on the chromatin apoptogenic agents, AIF translocates to the nucleus and
organization of NF-kB target loci. Signals other than DNA induces chromatin condensation and large-scale (50 kbp)
lesions, including steroid hormones, stress, and infection, may DNA fragmentation in a new form of caspase-independent
activate PARP molecules at specific chromosome sites. After apoptosis termed chromatinolysis.(68) Subsequent to its
modification with PAR, local chromatin proteins, transcription migration from mitochondria, AIF also triggers the release of
factors, and PARP-1 molecules themselves are proposed to mitochondrial cytochrome c and caspase activation. However,
dissociate from DNA and/or from pre-existing protein com- blockade of caspase activation does not abrogate AIF-
plexes, and to complex with nearby branched ADP-ribose dependent apoptotic cell death. How can PARP-1 induce
polymers. Consequently, the modified chromosome domain AIF translocation from the mitochondria to the nucleus?
may be transcribed and its chromatin constituents modified. The authors showed that the activity of PARP-1 is absolutely
Later on, due to the action of the poly (ADP-ribose) glyco- required for this process. The treatment of fibroblasts or
hydrolase (PARG) that cleaves PAR,(60) the chromatin would cortical neurons with very high doses of DNA-damaging
reform to return to the initial state. agents such as MNNG, NMDA or H2O2, induces cell death
Many questions still need to be answered, among them is through the translocation of AIF but the translocation is
the understanding of how PARP-1 is recruited to target genes inhibited in the presence of PARP inhibitor 3,4-dihydro-5-
and activated within normal, undamaged chromatin. Given [4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (DPQ) or in
that topoisomerase I is a necessary player in transcription and PARP-1-deficient cells.(67) AIF-neutralizing antibodies could
a partner of PARP-1, it is quite possible that, under a variety of block apoptosis but not caspases inhibitors despite the
circumstances, notably when transcription is particularly presence of features indicative of caspase-dependent cell
active as in puffs or in complex DNA sequence regions, death (such as cytochrome c release, caspase activation and
abortive topoI-SSB may arise leaving transient 50 DNA ends PARP-1 cleavage).(69) In addition, the overexpression of Bcl-
that particularly stimulate PARP-1.(61) A spontaneous PARP-1 2, one of the most powerful antiapoptotic proteins identified so
activation, as the one observed in Drosophila puffs, could well far, delayed but did not prevent PARP-1-dependent apoptosis.
be the result of transient topoisomerase I pauses. Recipro- Interestingly, it has been demonstrated that AIF is involved
cally, it has been recently shown that PAR formation at stalled in neuronal as well as myocardial cell death by ischemia-
topoisomerases I activates DNA break resealing.(62) reperfusion(70,71) in which PARP-1 plays a critical role. On the
Could these mechanisms described in the Drosophila be one hand, a very rapid signaling by nuclear PARP-1 to the
generalized to mammalians? If so what could be the role of mitochondria seems to cause AIF translocation. On the other
the homologues of PARP-1, like PARP-2, for which some hand, PAR accumulation in the central nervous system was
redundant functions have already been described? recently demonstrated in Drosophila mutants in which the
gene encoding the PAR degrading enzyme, poly(ADP-ribose)
The dark side of PARP-1: from the inflammatory glycohydrolase (PARG), was disrupted leading to progressive
response to AIF-mediated cell death neurodegeneration, reduced locomotion and short life-
The generation, by homologous recombination, of three span.(72) The key question that remains is which signal emitted
independent deficient mouse models have fully confirmed by the nucleus informs the mitochondria to release AIF?
the caretaker function of PARP-1 in mammalian cells Whatever the answer, AIF, as an essential downstream ef-
challenged by various genotoxins that directly or indirectly fector of PARP-1-mediated cell death, appears to be a future
break the DNA.(63–65) Unexpectedly, the knockout strategy therapeutic target in a number of inflammatory disorders.
revealed the instrumental role of PARP-1 in cell death in
various acute and chronic inflammatory disorders (see for Conclusions
review(8)). There has been increasing evidence that PARP-1 Most of the published data on PARP-1 and PARP-2 integrate
is, in fact, involved in the inflammatory response at two the unique property of these two DNA-dependent enzymes to
different levels: (i) activation of NF-kB and synthesis of detect DNA interruptions acting as potent enzymatic coacti-
proinflammatory factors(66) and (ii) increasing the sensitivity vators during various physiological or pathological processes.
of cells (particularly endothelial cells) to oxygen radicals They clearly participate in the first line of defense of the
produced during inflammation with consequences on DNA genome as efficient DNA-break sensors and signaling
damage, energy depletion and cell death. More recently, molecules as a part of a survival program. In contradiction,
Dawson and colleagues provided evidence that PARP-1 the response of a cell in which AIF has migrated from the
activity participates in the mechanism leading to cell death mitochondria to the nucleus is that the same enzymes now
through the release of a mitochondrial proapoptotic protein participate actively in cell death (Fig. 5). These two facets of
called apoptosis-inducing factor (AIF).(67) AIF is a flavoprotein PARP activity will undeniably continue to fascinate PARP

890 BioEssays 26.8


Review articles

Figure 5. The two facets of PARP activation. In replicating cells, limited damage to DNA induces PARP activity that allows the activation of
DNA repair pathways through the recruitment of DNA repair factors (i.e. XRCC1). The decision for the cell to engage the apoptotic pathway
after genotoxic stress takes place downstream of p53 activation, and the molecular determinants that switch between DNA repair and cell-
cycle arrest or apoptosis are not yet fully understood. As a consequence of the apoptosis option, PARPs are cleaved by caspases and are
thus inactivated to avoid futile repair. PARP inhibitors (red route) potentiate the cytotoxicity of DNA damaging agents. In post-mitotic cells,
reactive oxygen species (ROS) damage DNA, which in turn activates PARP and triggers AIF translocation to the nucleus.(67) The resulting
chromatinolysis exacerbates PARP activity and NAD depletion. The instrumentalization of PARP under these pathological conditions may
be responsible for the inflammatory status of the tissue/organ. PARP inhibitors are known to prevent both AIF translocation and
inflammatory injury.

aficionados for a while. PARP involvement in both cell survival 5. de Murcia G, Shall S, editors. 2000. From DNA damage and stress
and cell death gives rise to an intense industrial effort to signalling to cell death: poly (ADP-ribosylation) reactions. Oxford: Oxford
University Press; 238 p.
develop potent chemical inhibitors that, at the preclinical 6. Ruf A, Mennissier de Murcia J, de Murcia G, Schulz GE. 1996. Structure
phase, are now able to cause a complete regression of xeno- of the catalytic fragment of poly(AD-ribose) polymerase from chicken.
graft tumors.(73) Among the other members of the PARP Proc Natl Acad Sci USA 93:7481–7485.
7. Shall S, de Murcia G. 2000. Poly(ADP-ribose) polymerase-1: what have
family, the Tankyrase group, which is involved in telomere we learned from the deficient mouse model? Mutat Res 460:1–15.
functioning, and the MacroPARP group, whose function could 8. Burkle A. 2001. Physiology and pathophysiology of poly(ADP-ribosyl)a-
be associated with chromatin repression, may well constitute tion. Bioessays 23:795–806.
9. Shieh WM, Ame JC, Wilson MV, Wang ZQ, Koh DW, et al. 1998.
exciting fields of development for basic and applied science. Poly(ADP-ribose) polymerase null mouse cells synthesize ADP-ribose
polymers. J Biol Chem 273:30069–30072.
10. Ame JC, Rolli V, Schreiber V, Niedergang C, Apiou F, et al. 1999. PARP-
Acknowledgments 2, A novel mammalian DNA damage-dependent poly(ADP-ribose)
We thank all the members of the de Murcia laboratory and O. polymerase. J Biol Chem 274:17860–17868.
Poch for fruitful discussions. We also thank Dr. S. Natarajan- 11. Oliver AW, Ame JC, Roe SM, Good V, De Murcia G, et al. 2004. Crystal
structure of the catalytic fragment of murine poly(ADP-ribose) polymer-
Amé for careful reading of the manuscript. We are grateful to ase-2. Nucleic Acids Res 32:456–464.
Didier Hentsch (IGBMC, Illkirch), Jean-Christophe Laval 12. Poirier GG, de Murcia G, Jongstra-Bilen J, Niedergang C, Mandel P.
(Leica Microsystems, France) for their help with the laser 1982. Poly(ADP-ribosyl)ation of polynucleosomes causes relaxation of
chromatin structure. Proc Natl Acad Sci USA 79:3423–3427.
microbeam microscope. We thank Dr. Sugimura for the 10H 13. Aravind L, Koonin EV. 2000. SAP—a putative DNA-binding motif involved
anti-poly(ADP-ribose) antibody. in chromosomal organization. Trends Biochem Sci 25:112–114.
14. Dantzer F, Giraud-Panis MJ, Jaco I, Amé JC, Schultz I, et al. 2004.
Functional Interaction between Poly(ADP-Ribose) Polymerase 2 (PARP-
References 2) and TRF2: PARP Activity Negatively Regulates TRF2. Mol Cell Biol 24:
1. de Murcia G, Ménissier-de Murcia J. 1994. Poly(ADP-ribose) polymer- 1595–1607.
ase: a molecular nick-sensor. Trends Biochem. Sci 19:172–176. 15. Menissier de Murcia J, Ricoul M, Tartier L, Niedergang C, Huber A, et al.
2. D’Amours D, Desnoyers S, D’Silva I, Poirier GG. 1999. Poly(ADP- 2003. Functional interaction between PARP-1 and PARP-2 in chromo-
ribosyl)ation reactions in the regulation of nuclear functions. Biochem J some stability and embryonic development in mouse. Embo J 22:2255–
342:249–268. 2263.
3. Petrucco S. 2003. Sensing DNA damage by PARP-like fingers. Nucleic 16. Schreiber V, Ame JC, Dolle P, Schultz I, Rinaldi B, et al. 2002. Poly(ADP-
Acids Res 31:6689–6699. ribose) polymerase-2 (PARP-2) is required for efficient base excision
4. Kaufmann SH, Desnoyers S, Ottaviano Y, Davidson NE, Poirier GG. DNA repair in association with PARP-1 and XRCC1. J Biol Chem 277:
1993. Specific proteolytic cleavage of poly(ADP-ribose) polymerase: an 23028–23036.
early marker of chemotherapy-induced apoptosis. Cancer Res 53:3976– 17. Augustin A, Spenlehauer C, Dumond H, Menissier-De Murcia J, Piel M,
3985. et al. 2003. PARP-3 localizes preferentially to the daughter centriole and

BioEssays 26.8 891


Review articles

interferes with the G1/S cell cycle progression. J Cell Sci 116:1551– 41. Earle E, Saxena A, MacDonald A, Hudson DF, Shaffer LG, et al. 2000.
1562. Poly(ADP-ribose) polymerase at active centromeres and neocentro-
18. Kanai M, Tong WM, Sugihara E, Wang ZQ, Fukasawa K, et al. 2003. In- meres at metaphase. Hum Mol Genet 9:187–194.
volvement of poly(ADP-Ribose) polymerase 1 and poly(ADP-Ribosyl)- 42. Saxena A, Saffery R, Wong LH, Kalitsis P, Choo KH. 2002. Centromere
ation in regulation of centrosome function. Mol Cell Biol 23:2451–2462. proteins Cenpa, Cenpb, and Bub3 interact with poly(ADP-ribose)
19. Kickhoefer VA, Siva AC, Kedersha NL, Inman EM, Ruland C, et al. 1999. polymerase-1 protein and are poly(ADP-ribosyl)ated. J Biol Chem 277:
The 193-kD vault protein, VPARP, is a novel poly(ADP-ribose) poly- 26921–26926.
merase. J Cell Biol 146:917–928. 43. Saxena A, Wong LH, Kalitsis P, Earle E, Shaffer LG, et al. 2002.
20. Smith S, Giriat I, Schmitt A, de Lange T. 1998. Tankyrase, a poly(ADP- Poly(ADP-ribose) polymerase 2 localizes to mammalian active centro-
ribose) polymerase at human telomeres. Science 282:1484–1487. meres and interacts with PARP-1, Cenpa, Cenpb and Bub3, but not
21. De Rycker M, Venkatesan RN, Wei C, Price CM. 2003. Vertebrate Cenpc. Hum Mol Genet 11:2319–2329.
tankyrase domain structure and sterile alpha motif (SAM)-mediated 44. Catalan J, Surralles J, Falck GC, Autio K, Norppa H. 2000. Segregation
multimerization. Biochem J 372:87–96. of sex chromosomes in human lymphocytes. Mutagenesis 15:251–
22. Seimiya H, Muramatsu Y, Smith S, Tsuruo T. 2004. Functional subdomain 255.
in the ankyrin domain of Tankyrase 1 required for poly(ADP-ribosyl)ation 45. Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. 1998. DNA
of TRF1 and telomere elongation. Mol Cell Biol 24:1944–1955. double-stranded breaks induce histone H2AX phosphorylation on serine
23. Chi NW, Lodish HF. 2000. Tankyrase is a golgi-associated mitogen- 139. J Biol Chem 273:5858–5868.
activated protein kinase substrate that interacts with IRAP in GLUT4 46. Pleschke JM, Kleczkowska HE, Strohm M, Althaus FR. 2000. Poly(ADP-
vesicles. J Biol Chem 275:38437–38444. ribose) binds to specific domains in DNA damage checkpoint proteins.
24. Smith S, de Lange T. 2000. Tankyrase promotes telomere elongation in J Biol Chem 275:40974–40980.
human cells. Curr Biol 10:1299–1302. 47. Okano S, Lan L, Caldecott KW, Mori T, Yasui A. 2003. Spatial and
25. Chang W, Dynek JN, Smith S. 2003. TRF1 is degraded by ubiquitin- temporal cellular responses to single-strand breaks in human cells. Mol
mediated proteolysis after release from telomeres. Genes Dev 17:1328– Cell Biol 23:3974–3981.
1333. 48. Caldecott KW. 2003. XRCC1 and DNA strand break repair. DNA Repair
26. Dynek JN, Smith S. 2004. Resolution of sister telomere association is (Amst) 2:955–969.
required for progression through mitosis. Science 304:97–100. 49. Trucco C, Oliver FJ, de Murcia G, Menissier-de Murcia J. 1998. DNA
27. Kaminker PG, Kim SH, Taylor RD, Zebarjadian Y, Funk WD, et al. 2001. repair defect in poly(ADP-ribose) polymerase-deficient cell lines. Nucleic
TANK2, a new TRF1-associated poly(ADP-ribose) polymerase, causes Acids Res 26:2644–2649.
rapid induction of cell death upon overexpression. J Biol Chem 276: 50. Dantzer F, de La Rubia G, Menissier-De Murcia J, Hostomsky Z, de
35891–35899. Murcia G, et al. 2000. Base excision repair is impaired in mammalian
28. Cook BD, Dynek JN, Chang W, Shostak G, Smith S. 2002. Role for the cells lacking Poly(ADP-ribose) polymerase-1. Biochemistry 39:7559–
related poly(ADP-Ribose) polymerases tankyrase 1 and 2 at human 7569.
telomeres. Mol Cell Biol 22:332–342. 51. Lavrik OI, Prasad R, Sobol RW, Horton JK, Ackerman EJ, et al. 2001.
29. Lyons RJ, Deane R, Lynch DK, Ye ZS, Sanderson GM, et al. 2001. Photoaffinity labeling of mouse fibroblast enzymes by a base excision
Identification of a novel human tankyrase through its interaction with the repair intermediate. Evidence for the role of poly(ADP-ribose) polymer-
adaptor protein Grb14. J Biol Chem 276:17172–17180. ase-1 in DNA repair. J Biol Chem 276:25541–25548.
30. Bae J, Donigian JR, Hsueh AJ. 2003. Tankyrase 1 interacts with Mcl-1 52. Prasad R, Lavrik OI, Kim SJ, Kedar P, Yang XP, et al. 2001. DNA
proteins and inhibits their regulation of apoptosis. J Biol Chem 278: polymerase beta-mediated long patch base excision repair. Poly(ADP-
5195–5204. ribose)polymerase-1 stimulates strand displacement DNA synthesis.
31. Sbodio JI, Chi NW. 2002. Identification of a tankyrase-binding motif J Biol Chem 276:32411–32414.
shared by IRAP, TAB182, and human TRF1 but not mouse TRF1. NuMA 53. Leppard JB, Dong Z, Mackey ZB, Tomkinson AE. 2003. Physical and
contains this RXXPDG motif and is a novel tankyrase partner. J Biol functional interaction between DNA ligase IIIalpha and poly(ADP-
Chem 277:31887–31892. Ribose) polymerase 1 in DNA single-strand break repair. Mol Cell Biol
32. Sbodio JI, Lodish HF, Chi NW. 2002. Tankyrase-2 oligomerizes with 23:5919–5927.
tankyrase-1 and binds to both TRF1 (telomere-repeat-binding factor 1) 54. Fernandez-Capetillo O, Celeste A, Nussenzweig A. 2003. Focusing on
and IRAP (insulin-responsive aminopeptidase). Biochem J 361:451–459. foci: H2AX and the recruitment of DNA-damage response factors. Cell
33. Seimiya H, Smith S. 2002. The telomeric poly(ADP-ribose) polymerase, Cycle 2:426–427.
tankyrase 1, contains multiple binding sites for telomeric repeat binding 55. Bassing CH, Alt FW. 2004. H2AX May Function as an Anchor to Hold
factor 1 (TRF1) and a novel acceptor, 182-kDa tankyrase-binding protein Broken Chromosomal DNA Ends in Close Proximity. Cell Cycle 3:149–
(TAB182). J Biol Chem 277:14116–14126. 153.
34. Ma Q, Baldwin KT, Renzelli AJ, McDaniel A, Dong L. 2001. TCDD- 56. Tartier L, Spenlehauer C, Newman HC, Folkard M, Prise KM, et al. 2003.
inducible poly(ADP-ribose) polymerase: a novel response to 2,3,7,8- Local DNA damage by proton microbeam irradiation induces poly(ADP-
tetrachlorodibenzo-p-dioxin. Biochem Biophys Res Commun 289:499– ribose) synthesis in mammalian cells. Mutagenesis 18:411–416.
506. 57. Menissier-de Murcia J, Mark M, Wendling O, Wynshaw-Boris A, de
35. Bateman A, Birney E, Cerruti L, Durbin R, Etwiller L, et al. 2002. The Pfam Murcia G. 2001. Early embryonic lethality in PARP-1 Atm double-mutant
protein families database. Nucleic Acids Res 30:276–280. mice suggests a functional synergy in cell proliferation during develop-
36. Aravind L. 2001. The WWE domain: a common interaction module in ment. Mol Cell Biol 21:1828–1832.
protein ubiquitination and ADP ribosylation. Trends Biochem Sci 26:273– 58. Tulin A, Spradling A. 2003. Chromatin loosening by poly(ADP)-ribose
275. polymerase (PARP) at Drosophila puff loci. Science 299:560–562.
37. Aguiar RC, Yakushijin Y, Kharbanda S, Salgia R, Fletcher JA, et al. 2000. 59. Oliver FJ, Menissier-de Murcia J, Nacci C, Decker P, Andriantsitohaina
BAL is a novel risk-related gene in diffuse large B-cell lymphomas that R, et al. 1999. Resistance to endotoxic shock as a consequence of
enhances cellular migration. Blood 96:4328–4334. defective NF-kappaB activation in poly (ADP-ribose) polymerase-1
38. Allen MD, Buckle AM, Cordell SC, Lowe J, Bycroft M. 2003. The crystal deficient mice. Embo J 18:4446–4454.
structure of AF1521 a protein from Archaeoglobus fulgidus with homo- 60. Ame JC, Jacobson EL, Jacobson MK. 2000. ADP-ribose polymer
logy to the non-histone domain of macroH2A. J Mol Biol 330:503–511. metabolism. In: de Murcia G, Shall S, editors. From DNA Damage
39. Gagne JP, Shah RG, Poirier GG. 2001. Analysis of ADP-ribose polymer and Stress Signalling to Cell Death: Poly ADP-ribosylation Reactionss.
sizes in intact cells. Mol Cell Biochem 224:183–185. New-York: Oxford University Press; p 1–34.
40. Takada S, Kelkar A, Theurkauf WE. 2003. Drosophila checkpoint kinase 2 61. Pion E, Bombarda E, Stiegler P, Ullmann GM, Mely Y, et al. 2003.
couples centrosome function and spindle assembly to genomic integrity. Poly(ADP-ribose) polymerase-1 dimerizes at a 50 recessed DNA end
Cell 113:87–99. in vitro: a fluorescence study. Biochemistry 42:12409–12417.

892 BioEssays 26.8


Review articles

62. Malanga M, Althaus FR. 2004. Poly(ADP-ribose) reactivates stalled DNA 71. Zhu C, Qiu L, Wang X, Hallin U, Cande C, et al. 2003. Involvement of
topoisomerase I and Induces DNA strand break resealing. J Biol Chem apoptosis-inducing factor in neuronal death after hypoxia-ischemia in the
279:5244–5248. neonatal rat brain. J Neurochem 86:306–317.
63. Wang ZQ, Stingl L, Morrison C, Jantsch M, Los M, et al. 1997. PARP is 72. Hanai S, Kanai M, Ohashi S, Okamoto K, Yamada M, et al. 2004. Loss
important for genomic stability but dispensable in apoptosis. Genes Dev of poly(ADP-ribose) glycohydrolase causes progressive neurodegen-
11:2347–2358. eration in Drosophila melanogaster. Proc Natl Acad Sci USA 101:
64. Menissier de Murcia J, Niedergang C, Trucco C, Ricoul M, Dutrillaux B, 82–86.
et al. 1997. Requirement of poly(ADP-ribose)polymerase in recovery 73. Calabrese CR, Almassy R, Barton S, Batey MA, Calvert AH, et al. 2004.
from DNA damage in mice and in cells. Proc Natl Acad Sci USA 94: Anticancer chemosensitization and radiosensitization by the novel
7303–7307. poly(ADP-ribose) polymerase-1 inhibitor AG14361. J Natl Cancer Inst
65. Masutani M, Suzuki H, Kamada N, Watanabe M, Ueda O, et al. 1999. 96:56–67.
Poly(ADP-ribose) polymerase gene disruption conferred mice resistant to 74. Thompson JD, Higgins DG, Gibson TJ. 1994. CLUSTAL W: improving the
streptozotocin-induced diabetes. Proc Natl Acad Sci USA 96:2301–2304. sensitivity of progressive multiple sequence alignment through se-
66. Hassa PO, Hottiger MO. 1999. A role of poly (ADP-ribose) polymerase in quence weighting, position-specific gap penalties and weight matrix
NF-kappaB transcriptional activation. Biol Chem 380:953–959. choice. Nucleic Acids Res 22:4673–4680.
67. Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, et al. 2002. 75. Le Cam E, Fack F, Menissier-de Murcia J, Cognet JA, Barbin A, et al.
Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by 1994. Conformational analysis of a 139 base-pair DNA fragment
apoptosis-inducing factor. Science 297:259–263. containing a single-stranded break and its interaction with human
68. Penninger JM, Kroemer G. 2003. Mitochondria, AIF and caspases– poly(ADP-ribose) polymerase. J Mol Biol 235:1062–1071.
rivaling for cell death execution. Nat Cell Biol 5:97–99. 76. de Murcia G, Huletsky A, Poirier GG. 1988. Modulation of chromatin
69. Cande C, Vahsen N, Garrido C, Kroemer G. 2004. Apoptosis-inducing structure by poly(ADP-ribosyl)ation. Biochem Cell Biol 66:626–635.
factor (AIF): caspase-independent after all. Cell Death Differ. 77. Yie LY, Snow BE, Kickhoefer VA, Erdmann N, Zhou W, et al. 2004. Vault
70. Kim GT, Chun YS, Park JW, Kim MS. 2003. Role of apoptosis-inducing protein VPARP is associated with mammalian telomerase and is
factor in myocardial cell death by ischemia-reperfusion. Biochem dispensable for telomerase function and vault structure in vivo. Molec
Biophys Res Commun 309:619–624. Cell Biol, In Press.

BioEssays 26.8 893

You might also like