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.

  • Article
  • Published:

A common intronic variant of PARP1 confers melanoma risk and mediates melanocyte growth via regulation of MITF

Abstract

Previous genome-wide association studies have identified a melanoma-associated locus at 1q42.1 that encompasses a 100-kb region spanning the PARP1 gene. Expression quantitative trait locus (eQTL) analysis in multiple cell types of the melanocytic lineage consistently demonstrated that the 1q42.1 melanoma risk allele (rs3219090[G]) is correlated with higher PARP1 levels. In silico fine-mapping and functional validation identified a common intronic indel, rs144361550 (−/GGGCCC; r2 = 0.947 with rs3219090), as displaying allele-specific transcriptional activity. A proteomic screen identified RECQL as binding to rs144361550 in an allele-preferential manner. In human primary melanocytes, PARP1 promoted cell proliferation and rescued BRAFV600E-induced senescence phenotypes in a PARylation-independent manner. PARP1 also transformed TERT-immortalized melanocytes expressing BRAFV600E. PARP1-mediated senescence rescue was accompanied by transcriptional activation of the melanocyte-lineage survival oncogene MITF, highlighting a new role for PARP1 in melanomagenesis.

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: The melanoma risk-associated G allele at rs3219090 is correlated with increased PARP1 expression.
Figure 2: Functional annotation of a 3-kb region encompassing rs144361550 in primary melanocytes.
Figure 3: The melanoma-associated indel rs144361550 drives allelic transcriptional activity and protein binding.
Figure 4: RECQL binds to the insertion allele and mediates allelic expression.
Figure 5: Cell growth and H3K9me3 focus formation in primary human melanocytes expressing PARP1 and BRAFV600E.
Figure 6: MITF expression is restored in primary human melanocytes coexpressing PARP1 and BRAFV600E in a PARylation-independent manner, concurrent with partial reversal of senescence phenotypes.
Figure 7: PARP1 binds to the MITF-M promoter.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Amos, C.I. et al. Genome-wide association study identifies novel loci predisposing to cutaneous melanoma. Hum. Mol. Genet. 20, 5012–5023 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Barrett, J.H. et al. Genome-wide association study identifies three new melanoma susceptibility loci. Nat. Genet. 43, 1108–1113 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Barrett, J.H. et al. Fine mapping of genetic susceptibility loci for melanoma reveals a mixture of single variant and multiple variant regions. Int. J. Cancer 136, 1351–1360 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Bishop, D.T. et al. Genome-wide association study identifies three loci associated with melanoma risk. Nat. Genet. 41, 920–925 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Brown, K.M. et al. Common sequence variants on 20q11.22 confer melanoma susceptibility. Nat. Genet. 40, 838–840 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Iles, M.M. et al. A variant in FTO shows association with melanoma risk not due to BMI. Nat. Genet. 45, 428–432 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Law, M.H. et al. Genome-wide meta-analysis identifies five new susceptibility loci for cutaneous malignant melanoma. Nat. Genet. 47, 987–995 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Macgregor, S. et al. Genome-wide association study identifies a new melanoma susceptibility locus at 1q21.3. Nat. Genet. 43, 1114–1118 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rafnar, T. et al. Sequence variants at the TERTCLPTM1L locus associate with many cancer types. Nat. Genet. 41, 221–227 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Peña-Chilet, M. et al. Genetic variants in PARP1 (rs3219090) and IRF4 (rs12203592) genes associated with melanoma susceptibility in a Spanish population. BMC Cancer 13, 160 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Davies, J.R. et al. Inherited variation in the PARP1 gene and survival from melanoma. Int. J. Cancer 135, 1625–1633 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Law, M.H. et al. PARP1 polymorphisms play opposing roles in melanoma occurrence and survival. Int. J. Cancer 136, 2488–2489 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Krishnakumar, R. & Kraus, W.L. The PARP side of the nucleus: molecular actions, physiological outcomes, and clinical targets. Mol. Cell 39, 8–24 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Woodhouse, B.C. & Dianov, G.L. Poly ADP-ribose polymerase-1: an international molecule of mystery. DNA Repair (Amst.) 7, 1077–1086 (2008).

    Article  CAS  Google Scholar 

  15. 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).

    Article  CAS  Google Scholar 

  16. Bouchard, V.J., Rouleau, M. & Poirier, G.G. PARP-1, a determinant of cell survival in response to DNA damage. Exp. Hematol. 31, 446–454 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Maynard, S., Schurman, S.H., Harboe, C., de Souza-Pinto, N.C. & Bohr, V.A. Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 30, 2–10 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Swindall, A.F., Stanley, J.A. & Yang, E.S. PARP-1: friend or foe of DNA damage and repair in tumorigenesis? Cancers (Basel) 5, 943–958 (2013).

    Article  CAS  Google Scholar 

  19. Urabe, K. et al. The inherent cytotoxicity of melanin precursors: a revision. Biochim. Biophys. Acta 1221, 272–278 (1994).

    Article  CAS  PubMed  Google Scholar 

  20. Kuilman, T., Michaloglou, C., Mooi, W.J. & Peeper, D.S. The essence of senescence. Genes Dev. 24, 2463–2479 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Leikam, C., Hufnagel, A., Schartl, M. & Meierjohann, S. Oncogene activation in melanocytes links reactive oxygen to multinucleated phenotype and senescence. Oncogene 27, 7070–7082 (2008).

    Article  CAS  PubMed  Google Scholar 

  22. Kraus, W.L. Transcriptional control by PARP-1: chromatin modulation, enhancer-binding, coregulation, and insulation. Curr. Opin. Cell Biol. 20, 294–302 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Schiewer, M.J. & Knudsen, K.E. Transcriptional roles of PARP1 in cancer. Mol. Cancer Res. 12, 1069–1080 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hassa, P.O. & Hottiger, M.O. The functional role of poly(ADP-ribose) polymerase 1 as novel coactivator of NF-κB in inflammatory disorders. Cell. Mol. Life Sci. 59, 1534–1553 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Cervellera, M.N. & Sala, A. Poly(ADP-ribose) polymerase is a B-MYB coactivator. J. Biol. Chem. 275, 10692–10696 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  27. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Boersema, P.J., Raijmakers, R., Lemeer, S., Mohammed, S. & Heck, A.J. Multiplex peptide stable isotope dimethyl labeling for quantitative proteomics. Nat. Protoc. 4, 484–494 (2009).

    Article  CAS  PubMed  Google Scholar 

  29. Hubner, N.C., Nguyen, L.N., Hornig, N.C. & Stunnenberg, H.G. A quantitative proteomics tool to identify DNA–protein interactions in primary cells or blood. J. Proteome Res. 14, 1315–1329 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Butter, F. et al. Proteome-wide analysis of disease-associated SNPs that show allele-specific transcription factor binding. PLoS Genet. 8, e1002982 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Michaloglou, C. et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436, 720–724 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Kaplon, J. et al. A key role for mitochondrial gatekeeper pyruvate dehydrogenase in oncogene-induced senescence. Nature 498, 109–112 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Vredeveld, L.C. et al. Abrogation of BRAFV600E-induced senescence by PI3K pathway activation contributes to melanomagenesis. Genes Dev. 26, 1055–1069 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhuang, D. et al. c-MYC overexpression is required for continuous suppression of oncogene-induced senescence in melanoma cells. Oncogene 27, 6623–6634 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Garraway, L.A. et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 436, 117–122 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Eltze, T. et al. Imidazoquinolinone, imidazopyridine, and isoquinolindione derivatives as novel and potent inhibitors of the poly(ADP-ribose) polymerase (PARP): a comparison with standard PARP inhibitors. Mol. Pharmacol. 74, 1587–1598 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Wacker, D.A. et al. The DNA binding and catalytic domains of poly(ADP-ribose) polymerase 1 cooperate in the regulation of chromatin structure and transcription. Mol. Cell. Biol. 27, 7475–7485 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  39. Rolli, V., O'Farrell, M., Ménissier-de Murcia, J. & de Murcia, G. Random mutagenesis of the poly(ADP-ribose) polymerase catalytic domain reveals amino acids involved in polymer branching. Biochemistry 36, 12147–12154 (1997).

    Article  CAS  PubMed  Google Scholar 

  40. Haq, R. et al. Oncogenic BRAF regulates oxidative metabolism via PGC1α and MITF. Cancer Cell 23, 302–315 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Müller, J. et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat. Commun. 5, 5712 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Giuliano, S. et al. Microphthalmia-associated transcription factor controls the DNA damage response and a lineage-specific senescence program in melanomas. Cancer Res. 70, 3813–3822 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Huber, W.E. et al. A tissue-restricted cAMP transcriptional response: SOX10 modulates α-melanocyte-stimulating hormone-triggered expression of microphthalmia-associated transcription factor in melanocytes. J. Biol. Chem. 278, 45224–45230 (2003).

    Article  CAS  PubMed  Google Scholar 

  44. Takeda, K. et al. Induction of melanocyte-specific microphthalmia-associated transcription factor by Wnt-3a. J. Biol. Chem. 275, 14013–14016 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Ko, H.L. & Ren, E.C. Functional aspects of PARP1 in DNA repair and transcription. Biomolecules 2, 524–548 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Watanabe, A., Takeda, K., Ploplis, B. & Tachibana, M. Epistatic relationship between Waardenburg syndrome genes MITF and PAX3. Nat. Genet. 18, 283–286 (1998).

    Article  CAS  PubMed  Google Scholar 

  47. Lauss, M. et al. Genome-wide DNA methylation analysis in melanoma reveals the importance of CpG methylation in MITF regulation. J. Invest. Dermatol. 135, 1820–1828 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bochman, M.L., Paeschke, K. & Zakian, V.A. DNA secondary structures: stability and function of G-quadruplex structures. Nat. Rev. Genet. 13, 770–780 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Stefl, R. et al. A-like guanine–guanine stacking in the aqueous DNA duplex of d(GGGGCCCC). J. Mol. Biol. 307, 513–524 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Lu, X.J., Shakked, Z. & Olson, W.K. A-form conformational motifs in ligand-bound DNA structures. J. Mol. Biol. 300, 819–840 (2000).

    Article  CAS  PubMed  Google Scholar 

  51. Pollock, P.M. et al. High frequency of BRAF mutations in nevi. Nat. Genet. 33, 19–20 (2003).

    Article  CAS  PubMed  Google Scholar 

  52. Narita, M. et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell 113, 703–716 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Yokoyama, S. et al. A novel recurrent mutation in MITF predisposes to familial and sporadic melanoma. Nature 480, 99–103 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bertolotto, C. et al. A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma. Nature 480, 94–98 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Krishnakumar, R. et al. Reciprocal binding of PARP-1 and histone H1 at promoters specifies transcriptional outcomes. Science 319, 819–821 (2008).

    Article  CAS  PubMed  Google Scholar 

  56. 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).

    Article  CAS  PubMed  Google Scholar 

  57. Krishnakumar, R. & Kraus, W.L. PARP-1 regulates chromatin structure and transcription through a KDM5B-dependent pathway. Mol. Cell 39, 736–749 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Howie, B.N., Donnelly, P. & Marchini, J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet. 5, e1000529 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dignam, J.D., Lebovitz, R.M. & Roeder, R.G. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 11, 1475–1489 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The results appearing here are in part based on data generated by the TCGA Research Network (http://cancergenome.nih.gov/). Data were also obtained from the GTEx Portal on 2 December 2015 or dbGaP accession phs000424.v6.p1 on 17 December 2015. We would like to thank H. Widlund (Brigham and Women's Hospital, Harvard Medical School) for providing p'mel cells and technical assistance, D. Fisher (Dana-Farber/Harvard Cancer Center) for providing luciferase construct pMITF-382 and experimental advice, W. Pavan (National Human Genome Research Institute, National Institutes of Health) for providing luciferase construct pMITF-2256, M. Herlyn (Wistar Institute) for providing pLU-TCMV-FMCS-pPURO vector, G. Jönsson (Lund University) for assistance with assessment of MITF methylation, the Arizona State University DNA Laboratory, C. Hautman, C. Dagnall, K. Jones, and C. Chung at the National Cancer Institute Cancer Genomics Research Laboratory (CGR), D. Peeper (Netherlands Cancer Institute) for providing the lentiviral expression vector for BRAFV600E constructs, M. Webster and A. Weeraratna at the Wistar Institute Melanoma Research Center, S. Chanock, M. Dean, L. Amundadottir, J. Hoskins, L. Colli, A. Vu, and C. Lee from the National Cancer Institute, Laboratory of Translational Genomics, and D. Youngkin (Translational Genomics Research Institute). This work has been supported by the Intramural Research Program (IRP) of the Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health. The content of this publication does not necessarily reflect the views or policies of the US Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the US government. S.M. was supported by an Australian Research Council Future Fellowship, and N.K.H. was supported by a fellowship from the National Health and Medical Research Council of Australia. M.V. was supported by the Netherlands Organization for Scientific Research (NWO Gravitation Program Cancer Genomics Netherlands). M.M.M. was supported by a grant from the Marie Curie Initial Training Network (ITN) DevCom (FP7, grant 607142). M.M.I., J.A.N.-B., and D.T.B. were supported by the CRUK programme (c588/A19167) and the National Cancer Institute, National Institutes of Health (R01 CA083115). We would like to thank the GenoMEL consortium for its contributions.

Author information

Authors and Affiliations

Authors

Contributions

J.C., M.X., and K.M.B. designed the study. J.C., M.M.M., M.A.K., and W.J.K. conducted experiments for molecular characterization of PARP1 risk variants. M.X. performed phenotypic analyses of PARP1 in primary and immortalized melanocytes. Proteomics analysis was conducted by M.M.M. and M.V. In vitro biophysical analysis of DNA structures was performed by A.G. and M.-P.T.-F. Data were analyzed by T.Z., M.H.L., H.P., and M.M.I. Fine-mapping of GWAS data was performed by M.M.I., D.T.B., J.A.N.-B., S.M., and M.H.L. Melanoma cell line eQTL and ASE experiments were performed by K.M.B., N.K.H., J.M.T., M.G., and J.C. The manuscript was written by J.C., M.X., and K.M.B.

Corresponding author

Correspondence to Kevin M Brown.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–26, Supplementary Tables 1–7 and 9–14, and Supplementary Note. (PDF 23584 kb)

Supplementary Table 8

rs144361550 genotype of HapMap CEU individuals by fragment analysis. (XLSX 29 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Choi, J., Xu, M., Makowski, M. et al. A common intronic variant of PARP1 confers melanoma risk and mediates melanocyte growth via regulation of MITF. Nat Genet 49, 1326–1335 (2017). https://doi.org/10.1038/ng.3927

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3927

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