Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

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

Key Points

  • Poly(ADP-ribose) (PAR) is synthesized from NAD+ by PAR polymerases (PARPs) and regulates many physiological processes such as the maintenance of DNA integrity, gene expression and cell division.

  • PARPs form a superfamily of 17 members in humans, and display diverse subcellular distributions and functions. Some members might function together and possess overlapping properties.

  • PAR that is synthesized in response to DNA-strand breaks is a DNA-damage signalling molecule that allows a rapid and efficient cellular evaluation of the damage range. It is also an essential recruiting molecule that, in a few seconds, concentrates key factors of the single-strand break repair pathway at the site of the lesion.

  • The poly(ADP-ribosyl)ation of histones that are associated with open chromatin conformation at the DNA-damage site provided the first clue to the roles of PAR as an epigenetic modification. Recent evidence revealed an important role of PAR in the epigenetic regulation of chromatin structure and in gene expression under physiological conditions in which the integrity of the DNA is not affected.

  • The dogma that the DNA-damage-dependent PARP-1 is activated by DNA-strand breaks has to be reconsidered now due to recent studies that showed the activation of PARP-1 in the absence of DNA interruptions. Elucidating the triggers is currently one of our most exciting challenges.

  • PARP-1 and PAR play key roles in various acute and chronic inflammatory disorders as well as in a number of degenerative diseases by contributing to the caspase-independent, apoptosis-inducing factor (AIF)-dependent cell death. PARP inhibition confers protection to these pathologies.

  • PARP inhibitors have promising pharmacological applications in potentializing the effect of antitumour drugs in cancer therapy as well as in the treatment of inflammatory, neurological and cardiac disorders.

  • Emerging evidence indicates a possible functional interplay between the PAR metabolic pathway and the SIRT1-mediated deacetylation pathway in the regulation of chromatin structure and function that is associated with broad biological activities.

Abstract

The addition to proteins of the negatively charged polymer of ADP-ribose (PAR), which is synthesized by PAR polymerases (PARPs) from NAD+, is a unique post-translational modification. It regulates not only cell survival and cell-death programmes, but also an increasing number of other biological functions with which novel members of the PARP family have been associated. These functions include transcriptional regulation, telomere cohesion and mitotic spindle formation during cell division, intracellular trafficking and energy metabolism.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Metabolism of poly(ADP-ribose) during DNA damage and repair.
Figure 2: Structural alignment of PARP-1, PARP-2 and Corynebacterium diphtheriae toxin.
Figure 3: The PARP superfamily.
Figure 4: PARP-1 in the repair of single-strand breaks.
Figure 5: PARP at the crossroad of survival and inflammation.

Similar content being viewed by others

References

  1. Rongvaux, A., Andris, F., Van Gool, F. & Leo, O. Reconstructing eukaryotic NAD metabolism. Bioessays 25, 683–690 (2003).

    CAS  PubMed  Google Scholar 

  2. Berger, F., Ramirez-Hernandez, M. H. & Ziegler, M. The new life of a centenarian: signalling functions of NAD(P). Trends Biochem. Sci. 29, 111–118 (2004).

    CAS  PubMed  Google Scholar 

  3. Pleschke, J. M., Kleczkowska, H. E., Strohm, M. & Althaus, F. R. Poly(ADP-ribose) binds to specific domains in DNA damage checkpoint proteins. J. Biol. Chem. 275, 40974–40980 (2000). Identification of a PAR-binding motif that mediates selective interaction between PAR and protein partners.

    CAS  PubMed  Google Scholar 

  4. Ruf, A., Mennissier de Murcia, J., de Murcia, G. & Schulz, G. E. Structure of the catalytic fragment of poly(AD-ribose) polymerase from chicken. Proc. Natl Acad. Sci. USA 93, 7481–7485 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Oliver, A. W. et al. Crystal structure of the catalytic fragment of murine poly(ADP-ribose) polymerase-2. Nucleic Acids Res. 32, 456–464 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Amé, J. C., Spenlehauer, C. & de Murcia, G. The PARP superfamily. Bioessays 26, 882–893 (2004).

    PubMed  Google Scholar 

  7. Ma, Q., Baldwin, K. T., Renzelli, A. J., 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. 289, 499–506 (2001).

    CAS  PubMed  Google Scholar 

  8. Yu, M. et al. PARP-10, a novel Myc-interacting protein with poly(ADP-ribose) polymerase activity, inhibits transformation. Oncogene 24, 1982–1993 (2005).

    CAS  PubMed  Google Scholar 

  9. Aguiar, R. C., Takeyama, K., He, C., Kreinbrink, K. & Shipp, M. B-aggressive lymphoma (BAL) family proteins have unique domains that modulate transcription and exhibit poly(ADP-ribose) polymerase activity. J. Biol. Chem. 280, 33756–33765 (2005).

    CAS  PubMed  Google Scholar 

  10. Augustin, A. et al. PARP-3 localizes preferentially to the daughter centriole and interferes with the G1/S cell cycle progression. J. Cell Sci. 116, 1551–1562 (2003).

    CAS  PubMed  Google Scholar 

  11. Tulin, A., Stewart, D. & Spradling, A. C. The Drosophila heterochromatic gene encoding poly(ADP-ribose) polymerase (PARP) is required to modulate chromatin structure during development. Genes Dev. 16, 2108–2119 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Menissier de Murcia, J. et al. Requirement of poly(ADP-ribose) polymerase in recovery from DNA damage in mice and in cells. Proc. Natl Acad. Sci. USA 94, 7303–7307 (1997).

    CAS  Google Scholar 

  13. Wang, Z. Q. et al. PARP is important for genomic stability but dispensable in apoptosis. Genes Dev. 11, 2347–2358 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Masutani, M. et al. Poly(ADP-ribose) polymerase gene disruption conferred mice resistant to streptozotocin-induced diabetes. Proc. Natl Acad. Sci. USA 96, 2301–2304 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Jagtap, P. & Szabo, C. Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors. Nature Rev. Drug Discov. 4, 421–440 (2005).

    CAS  Google Scholar 

  16. Amé, J. C. et al. PARP-2, a novel mammalian DNA damage-dependent poly(ADP-ribose) polymerase. J. Biol. Chem. 274, 17860–17868 (1999).

    PubMed  Google Scholar 

  17. Menissier de Murcia, J. et al. Functional interaction between PARP-1 and PARP-2 in chromosome stability and embryonic development in mouse. EMBO J. 22, 2255–2263 (2003). Shows the partial redundancy between PARP-1 and PARP-2 in the maintenance of genome integrity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Schreiber, V. et al. 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. 277, 23028–23036 (2002).

    CAS  PubMed  Google Scholar 

  19. Gomez, M. et al. PARP-1 is a TRF2-associated poly(ADP-ribose) polymerase and protects eroded telomeres. Mol. Biol. Cell 17, 1686–1696 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Dantzer, F. et al. Functional interaction between poly(ADP-ribose) polymerase 2 (PARP-2) and TRF2: PARP activity negatively regulates TRF2. Mol. Cell. Biol. 24, 1595–1607 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Smith, S., Giriat, I., Schmitt, A. & de Lange, T. Tankyrase, a poly(ADP-ribose) polymerase at human telomeres. Science 282, 1484–1487 (1998).

    CAS  PubMed  Google Scholar 

  22. Sbodio, J. I. & Chi, N. W. Identification of a tankyrase-binding motif shared by IRAP, TAB182, and human TRF1 but not mouse TRF1. NuMA contains this RXXPDG motif and is a novel tankyrase partner. J. Biol. Chem. 277, 31887–31892 (2002).

    CAS  PubMed  Google Scholar 

  23. Sbodio, J. I., Lodish, H. F. & Chi, N. W. Tankyrase-2 oligomerizes with tankyrase-1 and binds to both TRF1 (telomere-repeat-binding factor 1) and IRAP (insulin-responsive aminopeptidase). Biochem. J. 361, 451–459 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Dynek, J. N. & Smith, S. Resolution of sister telomere association is required for progression through mitosis. Science 304, 97–100 (2004).

    CAS  PubMed  Google Scholar 

  25. Chang, P., Coughlin, M. & Mitchison, T. J. Tankyrase-1 polymerization of poly(ADP-ribose) is required for spindle structure and function. Nature Cell Biol. 7, 1133–1139 (2005).

    CAS  PubMed  Google Scholar 

  26. Hsiao, S. J., Poitras, M. F., Cook, B. D., Liu, Y. & Smith, S. Tankyrase 2 poly(ADP-ribose) polymerase domain-deleted mice exhibit growth defects but have normal telomere length and capping. Mol. Cell. Biol. 26, 2044–2054 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chiang, Y. J. et al. Generation and characterization of telomere length maintenance in tankyrase 2-deficient mice. Mol. Cell. Biol. 26, 2037–2043 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Matsuo, R., Murayama, A., Saitoh, Y., Sakaki, Y. & Inokuchi, K. Identification and cataloging of genes induced by long-lasting long-term potentiation in awake rats. J. Neurochem. 74, 2239–2249 (2000).

    CAS  PubMed  Google Scholar 

  29. Gao, G., Guo, X. & Goff, S. P. Inhibition of retroviral RNA production by ZAP, a CCCH-type zinc finger protein. Science 297, 1703–1706 (2002).

    CAS  PubMed  Google Scholar 

  30. Guo, X., Carroll, J. W., Macdonald, M. R., Goff, S. P. & Gao, G. The zinc finger antiviral protein directly binds to specific viral mRNAs through the CCCH zinc finger motifs. J. Virol. 78, 12781–12787 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ladurner, A. G. Inactivating chromosomes: a macro domain that minimizes transcription. Mol. Cell 12, 1–3 (2003).

    CAS  PubMed  Google Scholar 

  32. Karras, G. I. et al. The macro domain is an ADP-ribose binding module. EMBO J. 24, 1911–1920 (2005). Identification of the PAR-binding capacity of some macro domains

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kustatscher, G., Hothorn, M., Pugieux, C., Scheffzek, K. & Ladurner, A. G. Splicing regulates NAD metabolite binding to histone macroH2A. Nature Struct. Mol. Biol. 12, 624–625 (2005).

    CAS  Google Scholar 

  34. Goenka, S. & Boothby, M. Selective potentiation of Stat-dependent gene expression by collaborator of Stat6 (CoaSt6), a transcriptional cofactor. Proc. Natl Acad. Sci. USA 103, 4210–4215 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Aguiar, R. C. et al. BAL is a novel risk-related gene in diffuse large B-cell lymphomas that enhances cellular migration. Blood 96, 4328–4334 (2000).

    CAS  PubMed  Google Scholar 

  36. Kickhoefer, V. A. et al. The 193-kD vault protein, VPARP, is a novel poly(ADP-ribose) polymerase. J. Cell Biol. 146, 917–928 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, Y. et al. Vault poly(ADP-ribose) polymerase is associated with mammalian telomerase and is dispensable for telomerase function and vault structure in vivo. Mol. Cell. Biol. 24, 5314–5323 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Raval-Fernandes, S., Kickhoefer, V. A., Kitchen, C. & Rome, L. H. Increased susceptibility of vault poly(ADP-ribose) polymerase-deficient mice to carcinogen-induced tumorigenesis. Cancer Res. 65, 8846–8852 (2005).

    CAS  PubMed  Google Scholar 

  39. Chou, H. Y., Chou, H. T. & Lee, S. C. Cdk-dependent activation of poly(ADP-ribose) polymerase member 10 (PARP-10). J. Biol. Chem. 281, 15201–15207 (2006).

    CAS  PubMed  Google Scholar 

  40. Meyer-Ficca, M. L., Meyer, R. G., Coyle, D. L., Jacobson, E. L. & Jacobson, M. K. Human poly(ADP-ribose) glycohydrolase is expressed in alternative splice variants yielding isoforms that localize to different cell compartments. Exp. Cell Res. 297, 521–532 (2004).

    CAS  PubMed  Google Scholar 

  41. Koh, D. W. et al. Failure to degrade poly(ADP-ribose) causes increased sensitivity to cytotoxicity and early embryonic lethality. Proc. Natl Acad. Sci. USA 101, 17699–17704 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Hanai, S. et al. Loss of poly(ADP-ribose) glycohydrolase causes progressive neurodegeneration in Drosophila melanogaster. Proc. Natl Acad. Sci. USA 101, 82–86 (2004).

    CAS  PubMed  Google Scholar 

  43. Cortes, U. et al. Depletion of the 110-kilodalton isoform of poly(ADP-ribose) glycohydrolase increases sensitivity to genotoxic and endotoxic stress in mice. Mol. Cell. Biol. 24, 7163–7178 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Koh, D. W., Dawson, T. M. & Dawson, V. L. Poly(ADP-ribosyl)ation regulation of life and death in the nervous system. Cell. Mol. Life Sci. 62, 760–768 (2005).

    CAS  PubMed  Google Scholar 

  45. Yu, S. W. et al. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 297, 259–263 (2002). Demonstration that PARP-1 activation signals AIF release from mitochondria, resulting in a caspase-independent cell death.

    CAS  PubMed  Google Scholar 

  46. Oka, S., Kato, J. & Moss, J. Identification and characterization of a mammalian 39-kDa poly(ADP-ribose) glycohydrolase. J. Biol. Chem. 281, 705–713 (2006).

    CAS  PubMed  Google Scholar 

  47. Petrucco, S. Sensing DNA damage by PARP-like fingers. Nucleic Acids Res. 31, 6689–6699 (2003). PARP-1 zinc fingers belong to a protein family that is specialized in the detection of strand interruptions.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Poirier, G. G., de Murcia, G., Jongstra-Bilen, J., Niedergang, C. & Mandel, P. Poly(ADP-ribosyl)ation of polynucleosomes causes relaxation of chromatin structure. Proc. Natl Acad. Sci. USA 79, 3423–3427 (1982). The first demonstration of the relaxation of chromatin superstructure that is induced by poly(ADP-ribosyl)ation of histone H1.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Realini, C. A. & Althaus, F. R. Histone shuttling by poly(ADP-ribosylation). J. Biol. Chem. 267, 18858–18865 (1992).

    CAS  PubMed  Google Scholar 

  50. Masson, M. et al. XRCC1 is specifically associated with poly(ADP-ribose) polymerase and negatively regulates its activity following DNA damage. Mol. Cell. Biol. 18, 3563–3571 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Okano, S., Lan, L., Caldecott, K. W., Mori, T. & Yasui, A. Spatial and temporal cellular responses to single-strand breaks in human cells. Mol. Cell. Biol. 23, 3974–3981 (2003). Elegantly demonstrates the PAR-dependent recruitment of XRCC1 to a locally induced damaged site.

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Schreiber, V. et al. in Poly(ADP-ribosyl)ation Ch. 2 (ed. Burkle, A.) 13–31 (Landes Bioscience, Georgetown, 2006).

    Google Scholar 

  53. Caldecott, K. W. XRCC1 and DNA strand break repair. DNA Repair (Amst) 2, 955–969 (2003).

    CAS  Google Scholar 

  54. Trucco, C., Oliver, F. J., de Murcia, G. & Menissier- de Murcia, J. DNA repair defect in poly(ADP-ribose) polymerase-deficient cell lines. Nucleic Acids Res. 26, 2644–2649 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Heale, J. T. et al. Condensin I interacts with the PARP-1–XRCC1 complex and functions in DNA single-strand break repair. Mol. Cell 21, 837–848 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Yang, Y. G., Cortes, U., Patnaik, S., Jasin, M. & Wang, Z. Q. Ablation of PARP-1 does not interfere with the repair of DNA double-strand breaks, but compromises the reactivation of stalled replication forks. Oncogene 23, 3872–3882 (2004).

    CAS  PubMed  Google Scholar 

  57. Audebert, M., Salles, B. & Calsou, P. Involvement of poly(ADP-ribose) polymerase-1 and XRCC1/DNA ligase III in an alternative route for DNA double-strand breaks rejoining. J. Biol. Chem. 279, 55117–55126 (2004).

    CAS  PubMed  Google Scholar 

  58. Hochegger, H. et al. Parp-1 protects homologous recombination from interference by Ku and Ligase IV in vertebrate cells. EMBO J. 25, 1305–1314 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Menissier-de Murcia, J., Mark, M., Wendling, O., Wynshaw-Boris, A. & de Murcia, G. Early embryonic lethality in PARP-1 Atm double-mutant mice suggests a functional synergy in cell proliferation during development. Mol. Cell. Biol. 21, 1828–1832 (2001).

    Google Scholar 

  60. Henrie, M. S. et al. Lethality in PARP-1/Ku80 double mutant mice reveals physiological synergy during early embryogenesis. DNA Repair (Amst) 2, 151–158 (2003).

    CAS  Google Scholar 

  61. Huber, A., Bai, P., de Murcia, J. M. & de Murcia, G. PARP-1, PARP-2 and ATM in the DNA damage response: functional synergy in mouse development. DNA Repair (Amst) 3, 1103–1108 (2004).

    CAS  Google Scholar 

  62. Thiriet, C. & Hayes, J. J. Chromatin in need of a fix: phosphorylation of H2AX connects chromatin to DNA repair. Mol. Cell 18, 617–622 (2005).

    CAS  PubMed  Google Scholar 

  63. Tartier, L. et al. Local DNA damage by proton microbeam irradiation induces poly(ADP-ribose) synthesis in mammalian cells. Mutagenesis 18, 411–416 (2003).

    CAS  PubMed  Google Scholar 

  64. Du, Y. C. et al. The dynamic alterations of H2AX complex during DNA repair detected by a proteomic approach reveal the critical roles of Ca2+/calmodulin in the ionizing radiation induced cell cycle arrest. Mol. Cell. Proteomics 7 March 2006 (doi:10.1047/mcp.M500327-MCP200).

  65. Kruhlak, M. J. et al. Changes in chromatin structure and mobility in living cells at sites of DNA double-strand breaks. J. Cell Biol. 172, 823–834 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Slattery, E., Dignam, J. D., Matsui, T. & Roeder, R. G. Purification and analysis of a factor which suppresses nick-induced transcription by RNA polymerase II and its identity with poly(ADP-ribose) polymerase. J. Biol. Chem. 258, 5955–5959 (1983).

    CAS  PubMed  Google Scholar 

  67. Kim, M. Y., Mauro, S., Gevry, N., Lis, J. T. & Kraus, W. L. NAD+-dependent modulation of chromatin structure and transcription by nucleosome binding properties of PARP-1. Cell 119, 803–814 (2004). Shows the modulation of chromatin structure by PARP-1 and the activation of PARP-1 by nucleosomes in the absence of nicks.

    CAS  PubMed  Google Scholar 

  68. Tulin, A. & Spradling, A. Chromatin loosening by poly(ADP)-ribose polymerase (PARP) at Drosophila puff loci. Science 299, 560–562 (2003). Shows the requirement for PAR to loosen polytene-chromatin structure, which is associated with high gene expression.

    CAS  PubMed  Google Scholar 

  69. Kim, M. Y., Zhang, T. & Kraus, W. L. Poly(ADP-ribosyl)ation by PARP-1: 'PAR-laying' NAD+ into a nuclear signal. Genes Dev. 19, 1951–1967 (2005).

    CAS  PubMed  Google Scholar 

  70. Kraus, W. L. & Lis, J. T. PARP goes transcription. Cell 113, 677–683 (2003).

    CAS  PubMed  Google Scholar 

  71. Hassa, P. O., Buerki, C., Lombardi, C., Imhof, R. & Hottiger, M. O. Transcriptional coactivation of nuclear factor-κB-dependent gene expression by p300 is regulated by poly(ADP)-ribose polymerase-1. J. Biol. Chem. 278, 45145–45153 (2003).

    CAS  PubMed  Google Scholar 

  72. Oliver, F. J. et al. Resistance to endotoxic shock as a consequence of defective NF-κB activation in poly (ADP-ribose) polymerase-1 deficient mice. EMBO J. 18, 4446–4454 (1999). References 71 and 72 describe the co-activating function of PARP-1 in NF-κB-driven transcription of inflammatory genes.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Hassa, P. O. et al. Acetylation of poly(ADP-ribose) polymerase-1 by p300/CREB-binding protein regulates coactivation of NF-κB-dependent transcription. J. Biol. Chem. 280, 40450–40464 (2005).

    CAS  PubMed  Google Scholar 

  74. Pavri, R. et al. PARP-1 determines specificity in a retinoid signaling pathway via direct modulation of mediator. Mol. Cell 18, 83–96 (2005).

    CAS  PubMed  Google Scholar 

  75. Ju, B. G. et al. Activating the PARP-1 sensor component of the groucho–TLE1 corepressor complex mediates a CaMKinase IIδ-dependent neurogenic gene activation pathway. Cell 119, 815–829 (2004).

    CAS  PubMed  Google Scholar 

  76. Butler, A. J. & Ordahl, C. P. Poly(ADP-ribose) polymerase binds with transcription enhancer factor 1 to MCAT1 elements to regulate muscle-specific transcription. Mol. Cell. Biol. 19, 296–306 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Visochek, L. et al. PolyADP-ribosylation is involved in neurotrophic activity. J. Neurosci. 25, 7420–7428 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Midorikawa, R., Takei, Y. & Hirokawa, N. KIF4 motor regulates activity-dependent neuronal survival by suppressing PARP-1 enzymatic activity. Cell 125, 371–383 (2006).

    CAS  PubMed  Google Scholar 

  79. Cohen-Armon, M. et al. Long-term memory requires polyADP-ribosylation. Science 304, 1820–1822 (2004). Describes the necessity for PARP-1 activation during learning and the establishment of long-term memory in Aplysia.

    CAS  PubMed  Google Scholar 

  80. Lonskaya, I. et al. Regulation of poly(ADP-ribose) polymerase-1 by DNA structure-specific binding. J. Biol. Chem. 280, 17076–17083 (2005). Together with reference 81, a demonstration of PARP-1 activation by non DNA-break structures.

    CAS  PubMed  Google Scholar 

  81. Pion, E. et al. DNA-induced dimerization of poly(ADP-ribose) polymerase-1 triggers its activation. Biochemistry 44, 14670–14681 (2005).

    CAS  PubMed  Google Scholar 

  82. Maeda, Y. et al. PARP-2 interacts with TTF-1 and regulates expression of surfactant protein-B. J. Biol. Chem. 281, 9600–9606 (2006).

    CAS  PubMed  Google Scholar 

  83. Tulin, A., Naumova, N. M., Menon, A. K. & Spradling, A. C. Drosophila poly(ADP-ribose) glycohydrolase (Parg) mediates chromatin structure and Sir2-dependent silencing. Genetics 172, 363–371 (2005).

    PubMed  Google Scholar 

  84. Yu, W. et al. Poly(ADP-ribosyl)ation regulates CTCF-dependent chromatin insulation. Nature Genet. 36, 1105–1110 (2004).

    CAS  PubMed  Google Scholar 

  85. Saxena, A., Saffery, R., Wong, L. H., Kalitsis, P. & Choo, K. H. Centromere proteins Cenpa, Cenpb, and Bub3 interact with poly(ADP-ribose) polymerase-1 protein and are poly(ADP-ribosyl)ated. J. Biol. Chem. 277, 26921–26926 (2002).

    CAS  PubMed  Google Scholar 

  86. Saxena, A. et al. Poly(ADP-ribose) polymerase 2 localizes to mammalian active centromeres and interacts with PARP-1, Cenpa, Cenpb and Bub3, but not Cenpc. Hum. Mol. Genet. 11, 2319–2329 (2002).

    CAS  PubMed  Google Scholar 

  87. Monaco, L. et al. Inhibition of Aurora-B kinase activity by poly(ADP-ribosyl)ation in response to DNA damage. Proc. Natl Acad. Sci. USA 102, 14244–14248 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Fang, Y. et al. BubR1 is involved in regulation of DNA damage responses. Oncogene 30 January 2006 (doi:10.1038/sj.onc.1209392).

  89. Chang, P., Jacobson, M. K. & Mitchison, T. J. Poly(ADP-ribose) is required for spindle assembly and structure. Nature 432, 645–649 (2004).

    CAS  PubMed  Google Scholar 

  90. Oliver, F. J., Menissier-de Murcia, J. & de Murcia, G. Poly(ADP-ribose) polymerase in the cellular response to DNA damage, apoptosis, and disease. Am. J. Hum. Genet. 64, 1282–1288 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Burkart, V. et al. Mice lacking the poly(ADP-ribose) polymerase gene are resistant to pancreatic β-cell destruction and diabetes development induced by streptozocin. Nature Med. 5, 314–319 (1999).

    CAS  PubMed  Google Scholar 

  92. Mabley, J. G. et al. Anti-inflammatory effects of a novel, potent inhibitor of poly (ADP-ribose) polymerase. Inflamm. Res. 50, 561–569 (2001).

    CAS  PubMed  Google Scholar 

  93. Modjtahedi, N., Giordanetto, F., Madeo, F. & Kroemer, G. Apoptosis-inducing factor: vital and lethal. Trends Cell Biol. 16, 264–272 (2006).

    CAS  PubMed  Google Scholar 

  94. Wang, X., Yang, C., Chai, J., Shi, Y. & Xue, D. Mechanisms of AIF-mediated apoptotic DNA degradation in Caenorhabditis elegans. Science 298, 1587–1592 (2002).

    CAS  PubMed  Google Scholar 

  95. Zhang, J. Are poly(ADP-ribosyl)ation by PARP-1 and deacetylation by Sir2 linked? Bioessays 25, 808–814 (2003).

    CAS  PubMed  Google Scholar 

  96. Kruszewski, M. & Szumiel, I. Sirtuins (histone deacetylases III) in the cellular response to DNA damage — facts and hypotheses. DNA Repair (Amst) 4, 1306–1313 (2005).

    CAS  Google Scholar 

  97. Matsushita, N. et al. Role of NAD-dependent deacetylases SIRT1 and SIRT2 in radiation and cisplatin-induced cell death in vertebrate cells. Genes Cells 10, 321–332 (2005).

    CAS  PubMed  Google Scholar 

  98. Pillai, J. B., Isbatan, A., Imai, S. & Gupta, M. P. Poly(ADP-ribose) polymerase-1-dependent cardiac myocyte cell death during heart failure is mediated by NAD+ depletion and reduced Sir2α deacetylase activity. J. Biol. Chem. 280, 43121–43130 (2005).

    CAS  PubMed  Google Scholar 

  99. Araki, T., Sasaki, Y. & Milbrandt, J. Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science 305, 1010–1013 (2004).

    CAS  PubMed  Google Scholar 

  100. Curtin, N. J. PARP inhibitors for cancer therapy. Expert Rev. Mol. Med. 7, 1–20 (2005). Together with reference 15, excellent reviews describing the therapeutic promise of PARP inhibitors in cancer treatment or in inflammatory diseases.

    PubMed  Google Scholar 

  101. Calabrese, C. R. et al. Anticancer chemosensitization and radiosensitization by the novel poly(ADP-ribose) polymerase-1 inhibitor AG14361. J. Natl Cancer Inst. 96, 56–67 (2004).

    CAS  PubMed  Google Scholar 

  102. Farmer, H. et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434, 917–921 (2005).

    CAS  PubMed  Google Scholar 

  103. Bryant, H. E. et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434, 913–917 (2005). References 102 and 103 show the powerful capacity of PARP inhibitors to selectively kill BRCA2-deficient tumours.

    CAS  PubMed  Google Scholar 

  104. Hay, T. et al. Efficient deletion of normal Brca2-deficient intestinal epithelium by poly(ADP-ribose) polymerase inhibition models potential prophylactic therapy. Cancer Res. 65, 10145–10148 (2005).

    CAS  PubMed  Google Scholar 

  105. Kolthur-Seetharam, U., Dantzer, F., McBurney, M. W., de Murcia, G. & Sassone-Corsi, P. Control of AIF-mediated cell death by the functional interplay of SIRT1 and PARP-1 in response to DNA damage. Cell Cycle 5, 873–877 (2006).

    CAS  PubMed  Google Scholar 

  106. Bieganowski, P. & Brenner, C. Discoveries of nicotinamide riboside as a nutrient and conserved NRK genes establish a Preiss–Handler independent route to NAD+ in fungi and humans. Cell 117, 495–502 (2004).

    CAS  PubMed  Google Scholar 

  107. Kolisek, M., Beck, A., Fleig, A. & Penner, R. Cyclic ADP-ribose and hydrogen peroxide synergize with ADP-ribose in the activation of TRPM2 channels. Mol. Cell 18, 61–69 (2005).

    CAS  PubMed  Google Scholar 

  108. Bell, C. E. & Eisenberg, D. Crystal structure of diphtheria toxin bound to nicotinamide adenine dinucleotide. Adv. Exp. Med. Biol. 419, 35–43 (1997).

    CAS  PubMed  Google Scholar 

  109. Ruf, A., Rolli, V., de Murcia, G. & Schulz, G. E. The mechanism of the elongation and branching reaction of poly(ADP-ribose) polymerase as derived from crystal structures and mutagenesis. J. Mol. Biol. 278, 57–65 (1998).

    CAS  PubMed  Google Scholar 

  110. Ogata, N., Ueda, K. & Hayaishi, O. ADP-ribosylation of histone H2B. Identification of glutamic acid residue 2 as the modification site. J. Biol. Chem. 255, 7610–7615 (1980).

    CAS  PubMed  Google Scholar 

  111. Ogata, N., Ueda, K., Kagamiyama, H. & Hayaishi, O. ADP-ribosylation of histone H1. Identification of glutamic acid residues 2, 14, and the COOH-terminal lysine residue as modification sites. J. Biol. Chem. 255, 7616–7620 (1980).

    CAS  PubMed  Google Scholar 

  112. Khorasanizadeh, S. The nucleosome: from genomic organization to genomic regulation. Cell 116, 259–272 (2004).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the Centre National de la Recherche Scientifique, Association pour la Recherche contre le Cancer, Electricité de France, Ligue contre le Cancer, Commissariat à l'Energie Atomique and Agence Nationale pour la Recherche for their support. The authors also thank J. Ménissier-de Murcia for critical comments and suggestions on the manuscript and apologize to our colleagues for the omission of many pertinent references owing to space limitations.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Related links

Related links

FURTHER INFORMATION

Gilbert de Murcia's homepage

The PARP Link

National Center for Biotechnology Information

Glossary

Orthologues

Functionally related genes from different species that have evolved from the same ancestral gene.

Macro domain

A domain that constitutes the non-histone part of histone variant macroH2A. Some macro domains can bind ADP-ribose derivatives.

HMG proteins

A large protein family of small non-histone components of chromatin that function in higher-order chromatin structure.

Heterochromatin

A highly condensed and transcriptionally less active form of chromatin that is found at defined sites, such as centromeres, silencer DNA elements or telomeres.

Centromere

The region in eukaryote chromosomes where daughter chromatids are joined together, and on which the kinetochore assembles.

Centrosome

A structure that forms close to the nucleus in eukaryotic cells during interphase; it comprises a pair of centrioles, satellite bodies and a cytoplasmic zone, and, in animal cells, serves as the main microtubule-organizing centre.

WWE domain

A putative protein–protein interaction motif that contains two conserved Trp residues and one Glu residue.

CCCH zinc finger

A zinc-finger protein motif of the CX7–11CX3–9CX3H type that is a putative RNA-binding module.

Exosome

A complex of several exonucleases that functions in nuclei and the cytoplasm in several different RNA-processing and RNA-degradation pathways.

G1–S restriction point

Cells that progress through this cell-cycle checkpoint are committed to enter S phase.

SSBR/BER

(Single-strand break repair/base-excision repair). Repair pathways that are involved in the processing and repair of single-strand-DNA breaks that can arise indirectly as normal intermediates of DNA-base-excision repair or directly from the cleavage of the sugar–phosphate backbone.

Homologous recombination

A mechanism for the repair of double-strand-DNA breaks that uses a homologous double-stranded DNA molecule as a template for the repair of the broken DNA.

Non-homologous end joining

The predominant repair pathway that is used in mammals to repair double-strand-DNA breaks. No homology is required between the joined DNA strands.

Epigenetic regulation

The regulation of gene expression that occurs through transcriptional or post-transcriptional mechanisms, rather than the 'genetic' alteration of the genomic DNA sequence.

Mediator complex

A multiprotein complex that is recruited to some transcriptional enhancers by activator proteins.

Insulator

A chromatin boundary element that regulates gene activity in complex genetic loci by adopting a specialized chromatin structure and by preventing the interaction between enhancers and promoters.

Imprinted loci

The loci of a gene that is expressed from only one of the two parental copies. Which one is expressed is dependent on the sex of the parent from which the gene was derived.

Pericentric heterochromatin

A region of chromatin that is found adjacent to the centromere and that remains condensed throughout the cell cycle. It is considered to be typically constitutive heterochromatin.

Chromosomal passenger protein

A protein that associates with chromosomes during the early stages of mitosis, then with the spindle midzone and the equatorial cortex during anaphase and telophase, and with the midbody during exit from mitosis.

Spindle checkpoint

The molecular process that specifically controls the assembly of the kinetochore on the chromosomal centromere and the timing of kinetochore dissociation. Dissociation involves the movement of the kinetochores, along with their attached sister chromatids, to opposite poles of the mitotic spindle during anaphase.

Kinetochore

A large multiprotein complex that assembles onto the centromere of the chromosome and that links the chromosome to the microtubules of the mitotic spindle.

Chromatinolysis

The cleavage of DNA into high-molecular-weight (50 kb) fragments during caspase-independent cell death.

Mitochondrial membrane potential

An electrochemical gradient that occurs across the mitochondrial membrane, and is also referred to as ΔΨm. A loss in membrane potential serves as an early indicator of the initiation of cellular apoptosis.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Schreiber, V., Dantzer, F., Ame, JC. et al. Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol 7, 517–528 (2006). https://doi.org/10.1038/nrm1963

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm1963

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing