894
Views
5
CrossRef citations to date
0
Altmetric
Research Paper

DUSP3 maintains genomic stability and cell proliferation by modulating NER pathway and cell cycle regulatory proteins

ORCID Icon, ORCID Icon & ORCID Icon
Pages 1545-1561 | Received 17 Jan 2020, Accepted 20 Apr 2020, Published online: 07 May 2020

References

  • Kaliki S, Jajapuram SD, Maniar A, et al. Ocular and periocular tumors in xeroderma pigmentosum: A study of 120 Asian Indian patients. Am J Ophthalmol. 2019 Feb;198:146–153. doi:10.1016/j.ajo.2018.10.011. Epub 2018 Oct 16.
  • Mullenders LHF. Solar UV damage to cellular DNA: from mechanisms to biological effects. Photochem Photobiol Sci. 2018;17:1842–1852.
  • Weon JL, Glass DA II. Novel therapeutic approaches to xeroderma pigmentosum. Br J Dermatol. 2019 Aug;181(2):249–255. doi:10.1111/bjd.17253. Epub 2018 Nov 25.
  • Yoon JH, Prakash L, Prakash S. Highly error-free role of DNA polymerase eta in the replicative bypass of UV-induced pyrimidine dimers in mouse and human cells. Proc Natl Acad Sci U S A. 2009;106:18219–18224.
  • Hedglin M, Benkovic SJ. Regulation of Rad6/Rad18 activity during DNA damage tolerance. Annu Rev Biophys. 2015;44:207–228.
  • Hedglin M, Pandey B, Benkovic SJ. Characterization of human translesion DNA synthesis across a UV-induced DNA lesion. Elife. 2016 Oct 22;5. pii: e19788. doi:10.7554/eLife.19788.
  • Koch SC, Simon N, Ebert C, et al. Molecular mechanisms of xeroderma pigmentosum (XP) proteins. Q Rev Biophys. 2016;49:e5.
  • Rastogi RP, Richa KA, Tyagi MB, et al. Molecular mechanisms of ultraviolet radiation-induced DNA damage and repair. J Nucleic Acids. 2010;2010:592980.
  • Yu W, Li L, Wang G, et al. KU70 inhibition impairs both non-homologous end joining and homologous recombination DNA damage repair through SHP-1 induced dephosphorylation of SIRT1 in adult T-cell leukemia-lymphoma cells. Cell Physiol Biochem. 2018;49:2111–2123.
  • Kang Y, Cheong HM, Lee JH, et al. Protein phosphatase 5 is necessary for ATR-mediated DNA repair. Biochem Biophys Res Commun. 2011;404:476–481.
  • Nguyen TA, Slattery SD, Moon SH, et al. The oncogenic phosphatase WIP1 negatively regulates nucleotide excision repair. DNA Repair (Amst). 2010;9:813–823.
  • Torres TEP, Russo LC, Santos A, et al. Loss of DUSP3 activity radiosensitizes human tumor cell lines via attenuation of DNA repair pathways. Biochim Biophys Acta Gen Subj. 2017 Jul;1861(7):1879–1894. doi:10.1016/j.bbagen.2017.04.004. Epub 2017 Apr 4.
  • Alonso A, Sasin J, Bottini N, et al. Protein tyrosine phosphatases in the human genome. Cell. 2004;117:699–711.
  • Arnoldussen YJ, Lorenzo PI, Pretorius ME, et al. The mitogen-activated protein kinase phosphatase vaccinia H1-related protein inhibits apoptosis in prostate cancer cells and is overexpressed in prostate cancer. Cancer Res. 2008;68:9255–9264.
  • Chen YR, Chou HC, Yang CH, et al. Deficiency in VHR/DUSP3, a suppressor of focal adhesion kinase, reveals its role in regulating cell adhesion and migration. Oncogene. 2017;36:6509–6517.
  • Hoyt R, Zhu W, Cerignoli F, et al. Cutting edge: selective tyrosine dephosphorylation of interferon-activated nuclear STAT5 by the VHR phosphatase. J Immunol. 2007;179:3402–3406.
  • Rahmouni S, Cerignoli F, Alonso A, et al. Loss of the VHR dual-specific phosphatase causes cell-cycle arrest and senescence. Nat Cell Biol. 2006;8:524–531.
  • Panico K, Forti FL. Proteomic, cellular, and network analyses reveal new DUSP3 interactions with nucleolar proteins in HeLa cells. J Proteome Res. 2013;12:5851–5866.
  • Russo LC, Farias JO, Ferruzo PYM, et al. Revisiting the roles of VHR/DUSP3 phosphatase in human diseases. Clinics (Sao Paulo). 2018;73:e466s.
  • Monteiro LF, Ferruzo PYM, Russo LC, et al. DUSP3/VHR: a druggable dual phosphatase for human diseases. Rev Physiol Biochem Pharmacol. 2019;176:1–35. doi: 10.1007/112_2018_12.
  • Forti FL. Combined experimental and bioinformatics analysis for the prediction and identification of VHR/DUSP3 nuclear targets related to DNA damage and repair. Integr Biol (Camb). 2015;7:73–89.
  • Marteijn JA, Lans H, Vermeulen W, et al. Understanding nucleotide excision repair and its roles in cancer and ageing. Nat Rev Mol Cell Biol. 2014;15:465–481.
  • Lee JH, Hwang GS, Choi BS. Solution structure of a DNA decamer duplex containing the stable 3ʹ T.G base pair of the pyrimidine(6-4)pyrimidone photoproduct [(6-4) adduct]: implications for the highly specific 3ʹ T –> C transition of the (6-4) adduct. Proc Natl Acad Sci U S A. 1999;96:6632–6636.
  • McAteer K, Jing Y, Kao J, et al. Solution-state structure of a DNA dodecamer duplex containing a Cis-syn thymine cyclobutane dimer, the major UV photoproduct of DNA. J Mol Biol. 1998;282:1013–1032.
  • Cerignoli F, Rahmouni S, Ronai Z, et al. Regulation of MAP kinases by the VHR dual-specific phosphatase: implications for cell growth and differentiation. Cell Cycle. 2006;5:2210–2215.
  • Sugasawa K. Molecular mechanisms of DNA damage recognition for mammalian nucleotide excision repair. DNA Repair (Amst). 2016;44:110–117.
  • Russo LC, Minaya PY, Silva LE, et al. Assessing the roles of Rho GTPases in cell DNA repair by the nucleotide excision repair pathway. Methods Mol Biol. 2018;1821:319–338.
  • Hu J, Adebali O, Adar S, et al. Dynamic maps of UV damage formation and repair for the human genome. Proc Natl Acad Sci U S A. 2017;114:6758–6763.
  • Ryter SW, Tyrrell RM. Singlet molecular oxygen ((1)O2): a possible effector of eukaryotic gene expression. Free Radic Biol Med. 1998;24:1520–1534.
  • Cadet J, Sage E, Douki T. Ultraviolet radiation-mediated damage to cellular DNA. Mutat Res. 2005;571:3–17.
  • Besaratinia A, Yoon JI, Schroeder C, et al. Wavelength dependence of ultraviolet radiation-induced DNA damage as determined by laser irradiation suggests that cyclobutane pyrimidine dimers are the principal DNA lesions produced by terrestrial sunlight. Faseb J. 2011;25:3079–3091.
  • Chandrasekhar D, Van Houten B. In vivo formation and repair of cyclobutane pyrimidine dimers and 6-4 photoproducts measured at the gene and nucleotide level in Escherichia coli. Mutat Res. 2000;450:19–40.
  • Perdiz D, Grof P, Mezzina M, et al. Distribution and repair of bipyrimidine photoproducts in solar UV-irradiated mammalian cells. Possible role of Dewar photoproducts in solar mutagenesis. J Biol Chem. 2000;275:26732–26742.
  • Kobayashi N, Katsumi S, Imoto K, et al. Quantitation and visualization of ultraviolet-induced DNA damage using specific antibodies: application to pigment cell biology. Pigment Cell Res. 2001;14:94–102.
  • Riou L, Eveno E, van Hoffen A, et al. Differential repair of the two major UV-induced photolesions in trichothiodystrophy fibroblasts. Cancer Res. 2004;64:889–894.
  • Soufir N, Ged C, Bourillon A, et al. A prevalent mutation with founder effect in xeroderma pigmentosum group C from north Africa. J Invest Dermatol. 2010;130:1537–1542.
  • Satokata I, Tanaka K, Miura N, et al. Three nonsense mutations responsible for group A xeroderma pigmentosum. Mutat Res. 1992;273:193–202.
  • de Vries A, van Oostrom CT, Hofhuis FM, et al. Increased susceptibility to ultraviolet-B and carcinogens of mice lacking the DNA excision repair gene XPA. Nature. 1995;377:169–173.
  • Zhu Q, Wani AA. Nucleotide excision repair: finely tuned molecular orchestra of early pre-incision events. Photochem Photobiol. 2017;93:166–177.
  • Karikkineth AC, Scheibye-Knudsen M, Fivenson E, et al. Cockayne syndrome: clinical features, model systems and pathways. Ageing Res Rev. 2017;33:3–17.
  • Oghabi Bakhshaiesh T, Majidzadeh AK, Esmaeili R. Wip1: A candidate phosphatase for cancer diagnosis and treatment. DNA Repair (Amst). 2017;54:63–66.
  • Naderali E, Khaki AA, Rad JS, et al. Regulation and modulation of PTEN activity. Mol Biol Rep. 2018 Dec;45(6):2869–2881. doi:10.1007/s11033-018-4321-6. Epub 2018 Aug 25.
  • Huang CY, Hsieh FS, Wang CY, et al. Palbociclib enhances radiosensitivity of hepatocellular carcinoma and cholangiocarcinoma via inhibiting ataxia telangiectasia-mutated kinase-mediated DNA damage response. Eur J Cancer. 2018;102:10–22.
  • Carvalho H, Ortolan TG, dePaula T, et al. Sustained activation of p53 in confluent nucleotide excision repair-deficient cells resistant to ultraviolet-induced apoptosis. DNA Repair (Amst). 2008;7:922–931.
  • Zhi Y, Ji H, Pan J, et al. Downregulated XPA promotes carcinogenesis of bladder cancer via impairment of DNA repair. Tumour Biol. 2017;39:1010428317691679.
  • Ming P, Cai T, Li J, et al. A novel arylbenzofuran induces cervical cancer cell apoptosis and G1/S arrest through ERK-mediated Cdk2/cyclin-A signaling pathway. Oncotarget. 2016;7:41843–41856.
  • Kamenz J, Ferrell JE Jr. The Temporal Ordering Of Cell-Cycle Phosphorylation. Mol Cell. 2017;65:371–373.
  • Swaffer MP, Jones AW, Flynn HR, et al. CDK substrate phosphorylation and ordering the cell cycle. Cell. 2016;167:1750–61 e16.
  • Cundell MJ, Hutter LH, Nunes Bastos R, et al. A PP2A-B55 recognition signal controls substrate dephosphorylation kinetics during mitotic exit. J Cell Biol. 2016;214:539–554.
  • Goldberg ML. Cell cycle: abrogating interphase/M phase bistability. Curr Biol. 2018;28:R1342–R5.
  • Wigan M, Pinder A, Giles N, et al. A UVR-induced G2-phase checkpoint response to ssDNA gaps produced by replication fork bypass of unrepaired lesions is defective in melanoma. J Invest Dermatol. 2012;132:1681–1688.
  • Schmit TL, Ahmad N. Regulation of mitosis via mitotic kinases: new opportunities for cancer management. Mol Cancer Ther. 2007;6:1920–1931.
  • Gabrielli BG, Sarcevic B, Sinnamon J, et al. A cyclin D-Cdk4 activity required for G2 phase cell cycle progression is inhibited in ultraviolet radiation-induced G2 phase delay. J Biol Chem. 1999;274:13961–13969.
  • Jirawatnotai S, Hu Y, Livingston DM, et al. Proteomic identification of a direct role for cyclin d1 in DNA damage repair. Cancer Res. 2012;72:4289–4293.
  • Zhong Q, Hu Z, Li Q, et al. Cyclin D1 silencing impairs DNA double strand break repair, sensitizes BRCA1 wildtype ovarian cancer cells to olaparib. Gynecol Oncol. 2018;152:157–165.
  • Minella AC, Swanger J, Bryant E, et al. p53 and p21 form an inducible barrier that protects cells against cyclin E-cdk2 deregulation. Curr Biol. 2002;12:1817–1827.
  • Bendjennat M, Boulaire J, Jascur T, et al. UV irradiation triggers ubiquitin-dependent degradation of p21(WAF1) to promote DNA repair. Cell. 2003;114:599–610.
  • Al-Khalaf HH, Hendrayani SF, Aboussekhra A. ATR controls the p21(WAF1/Cip1) protein up-regulation and apoptosis in response to low UV fluences. Mol Carcinog. 2012;51:930–938.
  • Dotto GP. p21(WAF1/Cip1): more than a break to the cell cycle? Biochim Biophys Acta. 2000;1471:M43–56.
  • Itoh T, Linn S. The fate of p21CDKN1A in cells surviving UV-irradiation. DNA Repair (Amst). 2005;4:1457–1462.
  • Lehmann AR, Niimi A, Ogi T, et al. Translesion synthesis: Y-family polymerases and the polymerase switch. DNA Repair (Amst). 2007;6:891–899.
  • Soria G, Belluscio L, van Cappellen WA, et al. DNA damage induced Pol eta recruitment takes place independently of the cell cycle phase. Cell Cycle. 2009;8:3340–3348.
  • Gary R, Ludwig DL, Cornelius HL, et al. The DNA repair endonuclease XPG binds to proliferating cell nuclear antigen (PCNA) and shares sequence elements with the PCNA-binding regions of FEN-1 and cyclin-dependent kinase inhibitor p21. J Biol Chem. 1997;272:24522–24529.
  • Soria G, Speroni J, Podhajcer OL, et al. p21 differentially regulates DNA replication and DNA-repair-associated processes after UV irradiation. J Cell Sci. 2008;121:3271–3282.
  • Huschtscha LI, Holliday R. Limited and unlimited growth of SV40-transformed cells from human diploid MRC-5 fibroblasts. J Cell Sci. 1983;63:77–99.
  • Masutani C, Araki M, Yamada A, et al. Xeroderma pigmentosum variant (XP-V) correcting protein from HeLa cells has a thymine dimer bypass DNA polymerase activity. Embo J. 1999;18:3491–3501.
  • Espinha G, Osaki JH, Magalhaes YT, et al. Rac1 GTPase-deficient HeLa cells present reduced DNA repair, proliferation, and survival under UV or gamma irradiation. Mol Cell Biochem. 2015;404:281–297.
  • Magalhaes YT, Farias JO, Monteiro LF, et al. Measuring the contributions of the Rho Pathway to the DNA damage response in tumor epithelial cells. Methods Mol Biol. 2018;1821:339–355.
  • Russo LC, Araujo CB, Iwai LK, et al. A Cyclin D2-derived peptide acts on specific cell cycle phases by activating ERK1/2 to cause the death of breast cancer cells. J Proteomics. 2017;151:24–32.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.