Item request has been placed! ×
Item request cannot be made. ×
loading  Processing Request

Identification and characterization of mercaptopyrimidine-based small molecules as inhibitors of nonhomologous DNA end joining.

Item request has been placed! ×
Item request cannot be made. ×
loading   Processing Request
  • معلومة اضافية
    • المصدر:
      Publisher: Published by Blackwell Pub. on behalf of the Federation of European Biochemical Societies Country of Publication: England NLM ID: 101229646 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1742-4658 (Electronic) Linking ISSN: 1742464X NLM ISO Abbreviation: FEBS J Subsets: MEDLINE
    • بيانات النشر:
      Original Publication: Oxford, UK : Published by Blackwell Pub. on behalf of the Federation of European Biochemical Societies, c2005-
    • الموضوع:
    • نبذة مختصرة :
      Mercaptopyrimidine derivatives are heterocyclic compounds with potent biological activities including antiproliferative, antibacterial, and anti-inflammatory properties. The present study describes the synthesis and characterization of several mercaptopyrimidine derivatives through condensation of 5,6-diamino-2-mercaptopyrimidin-4-ol with various heterocyclic and aromatic aldehydes. Previous studies have shown that SCR7, synthesized from 5,6-diamino-2-mercaptopyrimidin-4-ol, induced cytotoxicity by targeting cancer cells by primarily inhibiting DNA Ligase IV involved in nonhomologous end joining, one of the major DNA double-strand break repair pathways. Inhibition of DNA repair pathways is considered as an important strategy for cancer therapy. Due to limitations of SCR7 in terms of IC 50 in cancer cells, here we have designed, synthesized, and characterized potent derivatives of SCR7 using 5,6-diamino-2-mercaptopyrimidin-4-ol as the starting material. Several synthesized imine compounds exhibited significant improvement in inhibition of end joining and cytotoxicity up to 27-fold lower concentrations than SCR7. Among these, two compounds, SCR116 and SCR132, showed increased cancer cell death in a Ligase IV-dependent manner. Treatment with the compounds also led to reduction in V(D)J recombination efficiency, cell cycle arrest at G2/M phase, accumulation of double-strand breaks inside cells, and improved anti-cancer potential when combined with γ-radiation and radiomimetic drugs. Thus, we describe novel inhibitors of NHEJ with higher efficacy and potential, which can be developed as cancer therapeutics.
      (© 2022 Federation of European Biochemical Societies.)
    • References:
      Ray U, Raghavan SC. Modulation of DNA double-strand break repair as a strategy to improve precise genome editing. Oncogene. 2020;39:6393-405.
      Srivastava M, Raghavan SC. DNA double-strand break repair inhibitors as cancer therapeutics. Chem Biol. 2015;22:17-29.
      Ghosh D, Raghavan SC. Nonhomologous end joining: new accessory factors fine tune the machinery. Trends Genet. 2021;37:582-99.
      Begg AC, Stewart FA, Vens C. Strategies to improve radiotherapy with targeted drugs. Nat Rev Cancer. 2011;11:239-53.
      Curtin NJ. DNA repair dysregulation from cancer driver to therapeutic target. Nat Rev Cancer. 2012;12:801-17.
      Darzynkiewicz Z, Traganos F, Wlodkowic D. Impaired DNA damage response - an Achilles' heel sensitizing cancer to chemotherapy and radiotherapy. Eur J Pharmacol. 2009;625:143-50.
      Trenner A, Sartori AA. Harnessing DNA double-Strand break repair for cancer treatment. Front Oncol. 2019;9:1388.
      Marangoni E, Foray N, O'Driscoll M, Douc-Rasy S, Bernier J, Bourhis J, et al. A Ku80 fragment with dominant negative activity imparts a radiosensitive phenotype to CHO-K1 cells. Nucleic Acids Res. 2000;28:4778-82.
      Ray U, Raghavan SC. Inhibitors of DNA double-strand break repair at the crossroads of cancer therapy and genome editing. Biochem Pharmacol. 2020;182:114195.
      Sishc BJ, Davis AJ. The role of the Core non-homologous end joining factors in carcinogenesis and cancer. Cancers. 2017;9:81.
      Cannan WJ, Pederson DS. Mechanisms and consequences of double-Strand DNA break formation in chromatin. J Cell Physiol. 2016;231:3-14.
      Jekimovs C, Bolderson E, Suraweera A, Adams M, O'Byrne KJ, Richard DJ. Chemotherapeutic compounds targeting the DNA double-strand break repair pathways: the good, the bad, and the promising. Front Oncol. 2014;4:86.
      Beskow C, Skikuniene J, Holgersson A, Nilsson B, Lewensohn R, Kanter L, et al. Radioresistant cervical cancer shows upregulation of the NHEJ proteins DNA-PKcs, Ku70 and Ku86. Br J Cancer. 2009;101:816-21.
      Kuschel B, Auranen A, McBride S, Novik KL, Antoniou A, Lipscombe JM, et al. Variants in DNA double-strand break repair genes and breast cancer susceptibility. Hum Mol Genet. 2002;11:1399-407.
      Pucci S, Mazzarelli P, Rabitti C, Giai M, Gallucci M, Flammia G, et al. Tumor specific modulation of KU70/80 DNA binding activity in breast and bladder human tumor biopsies. Oncogene. 2001;20:739-47.
      Shintani S, Mihara M, Li CN, Nakahara Y, Hino S, Nakashiro K, et al. Up-regulation of DNA-dependent protein kinase correlates with radiation resistance in oral squamous cell carcinoma. Cancer Sci. 2003;94:894-900.
      Sirzen F, Nilsson A, Zhivotovsky B, Lewensohn R. DNA-dependent protein kinase content and activity in lung carcinoma cell lines: correlation with intrinsic radiosensitivity. Eur J Cancer. 1999;35:111-6.
      Harnor SJ, Brennan A, Cano C. Targeting DNA-dependent protein kinase for cancer therapy. ChemMedChem. 2017;12:895-900.
      Mohiuddin IS, Kang MH. DNA-PK as an emerging therapeutic target in cancer. Front Oncol. 2019;9:635.
      Manjunath M, Choudhary B, Raghavan SC. SCR7, a potent cancer therapeutic agent and a biochemical inhibitor of nonhomologous DNA end-joining. Cancer Rep. 2021;4:e1341.
      Weterings E, Gallegos AC, Dominick LN, Cooke LS, Bartels TN, Vagner J, et al. A novel small molecule inhibitor of the DNA repair protein Ku70/80. DNA Repair. 2016;43:98-106.
      Ray U, Raul SK, Gopinatha VK, Ghosh D, Rangappa KS, Mantelingu K, et al. Identification and characterization of novel SCR7-based small-molecule inhibitor of DNA end-joining, SCR130 and its relevance in cancer therapeutics. Mol Carcinog. 2020;59:618-28.
      Ray U, Vartak SV, Raghavan SC. NHEJ inhibitor SCR7 and its different forms: promising CRISPR tools for genome engineering. Gene. 2020;763:144997.
      Srivastava M, Nambiar M, Sharma S, Karki SS, Goldsmith G, Hegde M, et al. An inhibitor of nonhomologous end-joining abrogates double-strand break repair and impedes cancer progression. Cell. 2012;151:1474-87.
      Vartak SV, Swarup HA, Gopalakrishnan V, Gopinatha VK, Ropars V, Nambiar M, et al. Autocyclized and oxidized forms of SCR7 induce cancer cell death by inhibiting nonhomologous DNA end joining in a ligase IV dependent manner. FEBS J. 2018;285:3959-76.
      Causse SZ, Marcion G, Chanteloup G, Uyanik B, Boudesco C, Grigorash BB, et al. HSP110 translocates to the nucleus upon genotoxic chemotherapy and promotes DNA repair in colorectal cancer cells. Oncogene. 2019;38:2767-77.
      Gkotzamanidou M, Terpos E, Bamia C, Munshi NC, Dimopoulos MA, Souliotis VL. DNA repair of myeloma plasma cells correlates with clinical outcome: the effect of the nonhomologous end-joining inhibitor SCR7. Blood. 2016;128:1214-25.
      Gopalakrishnan V, Gudapureddy R, Raghavan SC, Choudhary B. Inhibitor of nonhomologous end joining can inhibit proliferation of diffuse large B-cell lymphoma cells and potentiate the effect of ionization radiation. J Radiat Cancer Res. 2018;9:93-101.
      Kumar A, Bhatkar D, Jahagirdar D, Sharma NK. Non-homologous end joining inhibitor SCR-7 to exacerbate low-dose doxorubicin cytotoxicity in HeLa cells. J Cancer Prev. 2017;22:47-54.
      Chang HHY, Watanabe G, Gerodinnos CA, Ochi T, Blundell TL, Jackson SP, et al. Different DNA end configurations dictate which NHEJ components are Most important for joining efficiency. J Biol Chem. 2016;291:24377-89.
      John F, George J, Srivastava M, Hassan PA, Aswal VK, Karkie SS, et al. Pluronic copolymer encapsulated SCR7 as a potential anticancer agent. Faraday Discuss. 2015;177:155-61.
      John F, George J, Vartak SV, Srivastava M, Hassan PA, Aswal VK, et al. Enhanced efficacy of pluronic copolymer micelle encapsulated SCR7 against cancer cell proliferation. Macromol Biosci. 2015;15:521-34.
      Pandey M, Gopalakrishnan V, Swarup HA, Kumar S, Gudapureddy R, Jose AE, et al. Water-soluble version of SCR7-pyrazine inhibits DNA repair and abrogates tumor cell proliferation. J Radiat Cancer Res. 2019;10:27-43.
      Ray U, Jose AE, Suresh R, Kaloor U, Swarup HA, Nambiar M, et al. Water-soluble SCR7 can abrogate DNA end joining and induce cancer cell death. Clin Oncol Res. 2020;3:2-7.
      Lieber MR. The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem. 2010;79:181-211.
      Raghavan SC, Raman MJ. Nonhomologous end joining of complementary and noncomplementary DNA termini in mouse testicular extracts. DNA Repair. 2004;3:1297-310.
      Sathees CR, Raman MJ. Mouse testicular extracts process DNA double-strand breaks efficiently by DNA end-to-end joining. Mutat Res. 1999;433:1-13.
      Sharma S, Choudhary B, Raghavan SC. Efficiency of nonhomologous DNA end joining varies among somatic tissues, despite similarity in mechanism. Cell Mol Life Sci. 2011;68:661-76.
      Pandey M, Kumar S, Goldsmith G, Srivastava M, Elango S, Shameem M, et al. Identification and characterization of novel ligase I inhibitors. Mol Carcinog. 2017;56:550-66.
      Chiruvella KK, Sebastian R, Sharma S, Karande AA, Choudhary B, Raghavan SC. Time-dependent predominance of nonhomologous DNA end-joining pathways during embryonic development in mice. J Mol Biol. 2012;417:197-211.
      Kumar TS, Kari V, Choudhary B, Nambiar M, Akila TS, Raghavan SC. Anti-apoptotic protein BCL2 Down-regulates DNA end joining in cancer cells. J Biol Chem. 2010;285:32657-70.
      Grawunder U, Zimmer D, Fugmann S, Schwarz K, Lieber MR. DNA ligase IV is essential for V(D) J recombination and DNA double-Strand break repair in human precursor lymphocytes. Mol Cell. 1998;2:477-84.
      Jendele L, Krivak R, Skoda P, Novotny M, Hoksza D. PrankWeb: a web server for ligand binding site prediction and visualization. Nucleic Acids Res. 2019;47:W345-9.
      Ochi T, Gu XL, Blundell TL. Structure of the catalytic region of DNA ligase IV in complex with an Artemis fragment sheds light on double-Strand break repair. Structure. 2013;21:672-9.
      Nishana M, Raghavan SC. Role of recombination activating genes in the generation of antigen receptor diversity and beyond. Immunology. 2012;137:271-81.
      Gellert M. V(D) J recombination: RAG proteins, repair factors, and regulation. Annu Rev Biochem. 2002;71:101-32.
      Schatz DG, Swanson PC. V(D) J recombination: mechanisms of initiation. Annu Rev Genet. 2011;45:167-202.
      Raghavan SC, Lieber MR. Chromosomal translocations and non-B DNA structures in the human genome. Cell Cycle. 2004;3:762-8.
      Kumari R, Roy U, Desai S, Nilavar NM, Van Nieuwenhuijze A, Paranjape A, et al. MicroRNA miR-29c regulates RAG1 expression and modulates V(D) J recombination during B cell development. Cell Rep. 2021;36:109390.
      Raghavan SC, Tong J, Lieber MR. Hybrid joint formation in human V(D) J recombination requires nonhomologous DNA end joining. DNA Repair. 2006;5:278-85.
      Bunting SF, Callen E, Wong N, Chen HT, Polato F, Gunn A, et al. 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell. 2010;141:243-54.
      Gopalakrishnan V, Sharma S, Ray U, Manjunath M, Lakshmanan D, Vartak SV, et al. SCR7, an inhibitor of NHEJ can sensitize tumor cells to ionization radiation. Mol Carcinog. 2021;60:627-43.
      Chou T-C, Martin N. The mass-action law-based new computer software, CompuSyn, for automated simulation of synergism and antagonism in drug combination studies. Cancer Res. 2007;67:637-7.
      Azizi S, Mahdavi Shahri M, Rahman HS, Rahim RA, Rasedee A, Mohamad R. Green synthesis palladium nanoparticles mediated by white tea (Camellia sinensis) extract with antioxidant, antibacterial, and antiproliferative activities toward the human leukemia (MOLT-4) cell line. Int J Nanomedicine. 2017;12:8841-53.
      Zhao X, Loo SCJ, Lee PP-F, Tan TTY, Chu CK. Synthesis and cytotoxic activities of chloropyridylimineplatinum (II) and chloropyridyliminecopper (II) surface-functionalized poly (amidoamine) dendrimers. J Inorg Biochem. 2010;104:105-10.
      Keung MY, Wu Y, Badar F, Vadgama JV. Response of breast cancer cells to PARP inhibitors is independent of BRCA status. J Clin Med. 2020;9:940.
      Oplustil O'Connor L, Rulten SL, Cranston AN, Odedra R, Brown H, Jaspers JE, et al. The PARP inhibitor AZD2461 provides insights into the role of PARP3 inhibition for both synthetic lethality and tolerability with chemotherapy in preclinical models. Cancer Res. 2016;76:6084-94.
      Lai HC, Singh NP, Sasaki T. Development of artemisinin compounds for cancer treatment. Invest New Drugs. 2013;31:230-46.
      Diana P, Barraja P, Lauria A, Almerico AM, Cirrincione G, Loi AG, et al. 2-Triazenopyrroles: synthesis and biological activity. Eur J Med Chem. 1999;34:353-60.
      Yoshida K, Fujita A, Narazaki H, Asano T, Itoh Y. Drug resistance to nelarabine in leukemia cell lines might be caused by reduced expression of deoxycytidine kinase through epigenetic mechanisms. Cancer Chemother Pharmacol. 2022;89:83-91.
      Sharma A, Janocha AJ, Hill BT, Smith MR, Erzurum SC, Almasan A. Targeting mTORC1-mediated metabolic addiction overcomes Fludarabine resistance in malignant B cells. Mol Cancer Res. 2014;12:1205-15.
      Willmore E, de Caux S, Sunter NJ, Tilby MJ, Jackson GH, Austin CA, et al. A novel DNA-dependent protein kinase inhibitor, NU7026, potentiates the cytotoxicity of topoisomerase II poisons used in the treatment of leukemia. Blood. 2004;103:4659-65.
      Davidson D, Amrein L, Panasci L, Aloyz R. Small molecules, inhibitors of DNA-PK, targeting DNA repair, and beyond. Front Pharmacol. 2013;4:5.
      Zenke FT, Zimmermann A, Sirrenberg C, Dahmen H, Kirkin V, Pehl U, et al. Pharmacologic inhibitor of DNA-PK, M3814, potentiates radiotherapy and regresses human tumors in mouse models. Mol Cancer Ther. 2020;19:1091-101.
      Chen X, Zhong S, Zhu X, Dziegielewska B, Ellenberger T, Wilson GM, et al. Rational design of human DNA ligase inhibitors that target cellular DNA replication and repair. Cancer Res. 2008;68:3169-77.
      Shibata A, Jeggo P. A historical reflection on our understanding of radiation-induced DNA double strand break repair in somatic mammalian cells; interfacing the past with the present. Int J Radiat Biol. 2019;95:945-56.
      Davidson D, Coulombe Y, Martinez-Marignac VL, Amrein L, Grenier J, Hodkinson K, et al. Irinotecan and DNA-PKcs inhibitors synergize in killing of colon cancer cells. Invest New Drugs. 2012;30:1248-56.
      Veuger S, Curtin N, Richardson C, Smith G, Durkacz B. Radiosensitization and DNA repair inhibition by the combined use of novel inhibitors of DNA-dependent protein kinase and poly (ADP-ribose) polymerase-1. Cancer Res. 2003;63:6008-15.
      Ma W, Westmoreland J, Gordenin D, Resnick M. Alkylation Base damage is converted into repairable double-Strand breaks and complex intermediates in G2 cells lacking AP endonuclease. PLoS Genet. 2011;7:e1002059.
      Mehta A, Haber JE. Sources of DNA double-strand breaks and models of recombinational DNA repair. Cold Spring Harb Perspect Biol. 2014;6:a016428.
      Vartak SV, Raghavan SC. Inhibition of nonhomologous end joining to increase the specificity of CRISPR/Cas9 genome editing. FEBS J. 2015;282:4289-94.
      Roy U, Raghavan SC. Deleterious point mutations in T-cell acute lymphoblastic leukemia: mechanistic insights into leukemogenesis. Int J Cancer. 2021;149:1210-20.
      Chu VT, Weber T, Wefers B, Wurst W, Sander S, Rajewsky K, et al. Increasing the efficiency of homology-directed repair for CRISPR-Cas9-induced precise gene editing in mammalian cells. Nat Biotechnol. 2015;33:543-8.
      Maruyama T, Dougan SK, Truttmann MC, Bilate AM, Ingram JR, Ploegh HL. Increasing the efficiency of precise genome editing with CRISPR-Cas9 by inhibition of nonhomologous end joining. Nat Biotechnol. 2015;33:538-42.
      Singh P, Schimenti JC, Bolcun-Filas E. A mouse geneticist's practical guide to CRISPR applications. Genetics. 2015;199:1-15.
      Nambiar M, Raghavan SC. Mechanism of fragility at BCL2 gene minor breakpoint cluster region during t(14;18) chromosomal translocation. J Biol Chem. 2012;287:8688-701.
      Kumari N, Vartak SV, Dahal S, Kumari S, Desai SS, Gopalakrishnan V, et al. G-quadruplex structures contribute to differential Radiosensitivity of the human genome. iScience. 2019;21:288-307.
      Raghavan SC, Houston S, Hegde BG, Langen R, Haworth IS, Lieber MR. Stability and strand asymmetry in the non-B DNA structure at the bcl-2 major breakpoint region. J Biol Chem. 2004;279:46213-25.
      Sebastian R, Raghavan SC. Induction of DNA damage and erroneous repair can explain genomic instability caused by endosulfan. Carcinogenesis. 2016;37:929-40.
      Tripathi V, Agarwal H, Priya S, Batra H, Modi P, Pandey M, et al. MRN complex-dependent recruitment of ubiquitylated BLM helicase to DSBs negatively regulates DNA repair pathways. Nat Commun. 2018;9:1016.
      Kavitha CV, Nambiar M, Narayanaswamy PB, Thomas E, Rathore U, Kumar CSA, et al. Propyl-2-(8-(3,4-Difluorobenzyl)-2 ',5 '-Dioxo-8-Azaspiro [Bicyclo[3.2.1] Octane-3,4 '-Imidazolidine]-1 '-yl) acetate induces apoptosis in human leukemia cells through mitochondrial pathway following cell cycle arrest. PLoS One. 2013;8:e69103.
      Thomas E, Gopalakrishnan V, Hegde M, Kumar S, Karki SS, Raghavan SC, et al. A novel resveratrol based tubulin inhibitor induces mitotic arrest and activates apoptosis in cancer cells. Sci Rep. 2016;6, 34653.
      Vartak SV, Hegde M, Iyer D, Gaikwad S, Gopalakrishnan V, Srivastava M, et al. A novel inhibitor of BCL2, Disarib abrogates tumor growth while sparing platelets, by activating intrinsic pathway of apoptosis. Biochem Pharmacol. 2016;122:10-22.
      Iyer D, Vartak SV, Mishra A, Goldsmith G, Kumar S, Srivastava M, et al. Identification of a novel BCL2-specific inhibitor that binds predominantly to the BH1 domain. FEBS J. 2016;283:3408-37.
      Raghavan SC, Kirsch IR, Lieber MR. Analysis of the V(D) J recombination efficiency at lymphoid chromosomal translocation breakpoints*. J Biol Chem. 2001;276:29126-33.
      Raghavan SC, Swanson PC, Wu X, Hsieh C-L, Lieber MR. A non-B-DNA structure at the Bcl-2 major breakpoint region is cleaved by the RAG complex. Nature. 2004;428:88-93.
      Thomas E, Gopalakrishnan V, Somasagara RR, Choudhary B, Raghavan SC. Extract of Vernonia condensata, inhibits tumor progression and improves survival of tumor-allograft bearing mouse. Sci Rep. 2016;6, 23255.
      Dahal S, Dubey S, Raghavan SC. Homologous recombination-mediated repair of DNA double-strand breaks operates in mammalian mitochondria. Cell Mol Life Sci. 2018;75:1641-55.
      Chiruvella KK, Kari V, Choudhary B, Nambiar M, Ghanta RG, Raghavan SC. Methyl angolensate, a natural tetranortriterpenoid induces intrinsic apoptotic pathway in leukemic cells. FEBS Lett. 2008;582:4066-76.
      Shahabuddin MS, Nambiar M, Choudhary B, Advirao GM, Raghavan SC. A novel DNA intercalator, butylamino-pyrimido[4′,5′:4,5]selenolo(2,3-b) quinoline, induces cell cycle arrest and apoptosis in leukemic cells. Invest New Drugs. 2010;28:35-48.
    • Contributed Indexing:
      Keywords: DNA Ligase IV; NHEJ inhibitor; V(D)J recombination; cancer therapeutics; double-strand break repair; genomic instability
    • الرقم المعرف:
      9007-49-2 (DNA)
    • الموضوع:
      Date Created: 20220901 Date Completed: 20230203 Latest Revision: 20230222
    • الموضوع:
      20240513
    • الرقم المعرف:
      10.1111/febs.16615
    • الرقم المعرف:
      36048168