#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Using Existing Drugs as Leads for Broad Spectrum Anthelmintics Targeting Protein Kinases


As one of the largest protein families, protein kinases (PKs) regulate nearly all processes within the cell and are considered important drug targets. Much research has been conducted on inhibitors for PKs, leading to a wealth of compounds that target PKs that have potential to be lead anthelmintic drugs. Identifying compounds that have already been developed to treat neglected tropical diseases is an attractive way to obtain lead compounds inexpensively that can be developed into much needed drugs, especially for use in developing countries. In this study, PKs from nematodes, hosts, and DrugBank were identified and classified into kinase families and subfamilies. Nematode proteins were placed into orthologous groups that span the phylum Nematoda. A minimal kinome for the phylum Nematoda was identified, and properties of the minimal kinome were explored. Orthologous groups from the minimal kinome were prioritized for experimental testing based on RNAi phenotype of the Caenorhabditis elegans ortholog, transcript expression over the life-cycle and anatomic expression patterns. Compounds linked to targets in DrugBank belonging to the same kinase families and subfamilies in the minimal nematode kinome were extracted. Thirty-five compounds were tested in the non-parasitic C. elegans and active compounds progressed to testing against nematode species with different modes of parasitism, the blood-feeding Haemonchus contortus and the filarial Brugia malayi. Eighteen compounds showed efficacy in C. elegans, and six compounds also showed efficacy in at least one of the parasitic species. Hypotheses regarding the pathway the compounds may target and their molecular mechanism for activity are discussed.


Vyšlo v časopise: Using Existing Drugs as Leads for Broad Spectrum Anthelmintics Targeting Protein Kinases. PLoS Pathog 9(2): e32767. doi:10.1371/journal.ppat.1003149
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003149

Souhrn

As one of the largest protein families, protein kinases (PKs) regulate nearly all processes within the cell and are considered important drug targets. Much research has been conducted on inhibitors for PKs, leading to a wealth of compounds that target PKs that have potential to be lead anthelmintic drugs. Identifying compounds that have already been developed to treat neglected tropical diseases is an attractive way to obtain lead compounds inexpensively that can be developed into much needed drugs, especially for use in developing countries. In this study, PKs from nematodes, hosts, and DrugBank were identified and classified into kinase families and subfamilies. Nematode proteins were placed into orthologous groups that span the phylum Nematoda. A minimal kinome for the phylum Nematoda was identified, and properties of the minimal kinome were explored. Orthologous groups from the minimal kinome were prioritized for experimental testing based on RNAi phenotype of the Caenorhabditis elegans ortholog, transcript expression over the life-cycle and anatomic expression patterns. Compounds linked to targets in DrugBank belonging to the same kinase families and subfamilies in the minimal nematode kinome were extracted. Thirty-five compounds were tested in the non-parasitic C. elegans and active compounds progressed to testing against nematode species with different modes of parasitism, the blood-feeding Haemonchus contortus and the filarial Brugia malayi. Eighteen compounds showed efficacy in C. elegans, and six compounds also showed efficacy in at least one of the parasitic species. Hypotheses regarding the pathway the compounds may target and their molecular mechanism for activity are discussed.


Zdroje

1. CohenP (2001) The role of protein phosphorylation in human health and disease. The Sir Hans Krebs Medal Lecture. Eur J Biochem 268: 5001–5010.

2. BoyleSN, KoleskeAJ (2007) Dissecting kinase signaling pathways. Drug Discov Today 12: 717–724.

3. CohenP (2002) Protein kinases–the major drug targets of the twenty-first century? Nat Rev Drug Discov 1: 309–315.

4. ManningG (2005) Genomic overview of protein kinases. WormBook 1–19.

5. ZhangJ, YangPL, GrayNS (2009) Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer 9: 28–39.

6. CohenP (2000) The regulation of protein function by multisite phosphorylation–a 25 year update. Trends Biochem Sci 25: 596–601.

7. Miranda-SaavedraD, BartonGJ (2007) Classification and functional annotation of eukaryotic protein kinases. Proteins 68: 893–914.

8. Holden-DyeL, WalkerRJ (2007) Anthelmintic drugs. WormBook 1–13.

9. PrichardRK, GearyTG (2008) Drug discovery: fresh hope to can the worms. Nature 452: 157–158.

10. van den EndenE (2009) Pharmacotherapy of helminth infection. Expert Opin Pharmacother 10: 435–451.

11. DoerigC (2004) Protein kinases as targets for anti-parasitic chemotherapy. Biochim Biophys Acta 1697: 155–168.

12. PollastriMP, CampbellRK (2011) Target repurposing for neglected diseases. Future Med Chem 3: 1307–1315.

13. SundarS, JhaTK, ThakurCP, EngelJ, SindermannH, et al. (2002) Oral miltefosine for Indian visceral leishmaniasis. N Engl J Med 347: 1739–1746.

14. PepinJ, MilordF, GuernC, SchechterPJ (1987) Difluoromethylornithine for arseno-resistant Trypanosoma brucei gambiense sleeping sickness. Lancet 2: 1431–1433.

15. GhedinE, WangS, SpiroD, CalerE, ZhaoQ, et al. (2007) Draft genome of the filarial nematode parasite Brugia malayi. Science 317: 1756–1760.

16. AbadP, GouzyJ, AuryJM, Castagnone-SerenoP, DanchinEG, et al. (2008) Genome sequence of the metazoan plant-parasitic nematode Meloidogyne incognita. Nat Biotechnol 26: 909–915.

17. MitrevaM, JasmerDP, ZarlengaDS, WangZ, AbubuckerS, et al. (2011) The draft genome of the parasitic nematode Trichinella spiralis. Nat Genet 43: 228–235.

18. BrownleeDJ, Holden-DyeL, WalkerRJ (1997) Actions of the anthelmintic ivermectin on the pharyngeal muscle of the parasitic nematode, Ascaris suum. Parasitology 115: 553–561.

19. JasmerDP, YaoC, RehmanA, JohnsonS (2000) Multiple lethal effects induced by a benzimidazole anthelmintic in the anterior intestine of the nematode Haemonchus contortus. Molecular and Biochemical Parasitology 105: 81–90.

20. CharvetCL, RobertsonAP, CabaretJ, MartinRJ, NeveuC (2012) Selective effect of the anthelmintic bephenium on Haemonchus contortus levamisole-sensitive acetylcholine receptors. Invert Neurosci 12: 43–51.

21. BrindleyPJ, MitrevaM, GhedinE, LustigmanS (2009) Helminth genomics: The implications for human health. PLoS Negl Trop Dis 3: e538.

22. KwokTC, RickerN, FraserR, ChanAW, BurnsA, et al. (2006) A small-molecule screen in C. elegans yields a new calcium channel antagonist. Nature 441: 91–95.

23. MortonS, DavisRJ, CohenP (2004) Signalling pathways involved in multisite phosphorylation of the transcription factor ATF-2. FEBS Lett 572: 177–183.

24. KamathA, WangJ, LeeF, MaratheP (2008) Preclinical pharmacokinetics and in vitro metabolism of dasatinib (BMS-354825): a potent oral multi-targeted kinase inhibitor against SRC and BCR-ABL. Cancer Chemotherapy and Pharmacology 61: 365–376.

25. ChristopherLJ, CuiD, LiW, BarrosA, AroraVK, et al. (2008) Biotransformation of [14C]Dasatinib: In Vitro Studies in Rat, Monkey, and Human and Disposition after Administration to Rats and Monkeys. Drug Metabolism and Disposition 36: 1341–1356.

26. ZhaiS, SenderowiczA, SausvilleE, FiggW (2002) Flavopiridol, a novel cyclin-dependent kinase inhibitor, in clinical development. The Annals of Pharmacotherapy 36: 905–911.

27. SenderowiczAM, HeadleeD, StinsonSF, LushRM, KalilN, et al. (1998) Phase I trial of continuous infusion flavopiridol, a novel cyclin-dependent kinase inhibitor, in patients with refractory neoplasms. Journal of Clinical Oncology 16: 2986–2999.

28. ShapiroGI, SupkoJG, PattersonA, LynchC, LuccaJ, et al. (2001) A Phase II Trial of the Cyclin-dependent Kinase Inhibitor Flavopiridol in Patients with Previously Untreated Stage IV Non-Small Cell Lung Cancer. Clinical Cancer Research 7: 1590–1599.

29. ShaikhB, JacksonJ, GuyerG, RavisWR (1991) Determination of neomycin in plasma and urine by high-performance liquid chromatography. Application to a preliminary pharmacokinetic study. J Chromatogr 571: 189–198.

30. ZhuoX, ZhengN, FelixCA, BlairIA (2004) KINETICS AND REGULATION OF CYTOCHROME P450-MEDIATED ETOPOSIDE METABOLISM. Drug Metabolism and Disposition 32: 993–1000.

31. KishiS, YangW, BoureauB, MorandS, DasS, et al. (2004) Effects of prednisone and genetic polymorphisms on etoposide disposition in children with acute lymphoblastic leukemia. Blood 103: 67–72.

32. BrooksDJ, SrinivasNR, AlbertsDS, ThomasT, IgwemzieLM, et al. (1995) Phase I and pharmacokinetic study of etoposide phosphate. Anticancer Drugs 6: 637–644.

33. ArbuckSG, DouglassHO, CromWR, GoodwinP, SilkY, et al. (1986) Etoposide pharmacokinetics in patients with normal and abnormal organ function. Journal of Clinical Oncology 4: 1690–1695.

34. PaoliniGV, ShaplandRH, van HoornWP, MasonJS, HopkinsAL (2006) Global mapping of pharmacological space. Nat Biotechnol 24: 805–815.

35. DavisMI, HuntJP, HerrgardS, CiceriP, WodickaLM, et al. (2011) Comprehensive analysis of kinase inhibitor selectivity. Nat Biotechnol 29: 1046–1051.

36. WeiS, MarchesF, DanielB, SondaS, HeidenreichK, et al. (2002) Pyridinylimidazole p38 mitogen-activated protein kinase inhibitors block intracellular Toxoplasma gondii replication. Int J Parasitol 32: 969–977.

37. WeiS, DanielBJ, BrumlikMJ, BurowME, ZouW, et al. (2007) Drugs designed to inhibit human p38 mitogen-activated protein kinase activation treat Toxoplasma gondii and Encephalitozoon cuniculi infection. Antimicrob Agents Chemother 51: 4324–4328.

38. GelmedinV, Caballero-GamizR, BrehmK (2008) Characterization and inhibition of a p38-like mitogen-activated protein kinase (MAPK) from Echinococcus multilocularis: antiparasitic activities of p38 MAPK inhibitors. Biochem Pharmacol 76: 1068–1081.

39. BrumlikMJ, NkhomaS, KiousMJ, ThompsonGR3rd, PattersonTF, et al. (2011) Human p38 mitogen-activated protein kinase inhibitor drugs inhibit Plasmodium falciparum replication. Exp Parasitol 128: 170–175.

40. LiB-W, WangZ, RushAC, MitrevaM, WeilGJ (2012) Transcription profiling reveals stage-and function-dependent expression patterns in the filarial nematode Brugia malayi. BMC Genomics 13: 184.

41. EglenRM, ReisineT (2009) The current status of drug discovery against the human kinome. Assay Drug Dev Technol 7: 22–43.

42. CheongJK, VirshupDM (2011) Casein kinase 1: Complexity in the family. Int J Biochem Cell Biol 43: 465–469.

43. UrbaniakMD (2009) Casein kinase 1 isoform 2 is essential for bloodstream form Trypanosoma brucei. Mol Biochem Parasitol 166: 183–185.

44. KnockaertM, GrayN, DamiensE, ChangYT, GrellierP, et al. (2000) Intracellular targets of cyclin-dependent kinase inhibitors: identification by affinity chromatography using immobilised inhibitors. Chem Biol 7: 411–422.

45. CalabokisM, KurzL, GonzattiMI, BubisJ (2003) Protein kinase CK1 from Trypanosoma cruzi. J Protein Chem 22: 591–599.

46. FongDH, XiongB, HwangJ, BerghuisAM (2011) Crystal structures of two aminoglycoside kinases bound with a eukaryotic protein kinase inhibitor. PLoS One 6: e19589.

47. MurrayP, CleggRA, ReesHH, FisherMJ (2008) siRNA-mediated knockdown of a splice variant of the PK-A catalytic subunit gene causes adult-onset paralysis in C. elegans. Gene 408: 157–163.

48. DongM, YanBP, LiaoJK, LamYY, YipGW, et al. (2010) Rho-kinase inhibition: a novel therapeutic target for the treatment of cardiovascular diseases. Drug Discov Today 15: 622–629.

49. TaoufiqZ, GayF, BalvanyosJ, CiceronL, TefitM, et al. (2008) Rho kinase inhibition in severe malaria: thwarting parasite-induced collateral damage to endothelia. J Infect Dis 197: 1062–1073.

50. FischerP, DjohaS, ButtnerDW, ZipfelPF (2003) Isolation and characterization of the regulatory subunit of cAMP-dependent protein kinase from the filarial parasite Onchocerca volvulus. Mol Biochem Parasitol 128: 33–42.

51. WurtzN, ChapusC, DesplansJ, ParzyD (2011) cAMP-dependent protein kinase from Plasmodium falciparum: an update. Parasitology 138: 1–25.

52. LiL, StoeckertCJJr, RoosDS (2003) OrthoMCL: identification of ortholog groups for eukaryotic genomes. Genome Res 13: 2178–2189.

53. MartinJ, AbubuckerS, HeizerE, TaylorCM, MitrevaM (2012) Nematode.net update 2011: addition of data sets and tools featuring next-generation sequencing data. Nucleic Acids Res 40: D720–728.

54. KumarS, ChaudharyK, FosterJM, NovelliJF, ZhangY, et al. (2007) Mining predicted essential genes of Brugia malayi for nematode drug targets. PLoS One 2: e1189.

55. TaylorCM, FischerK, AbubuckerS, WangZ, MartinJ, et al. (2011) Targeting protein-protein interactions for parasite control. PLoS One 6: e18381.

56. YookK, HarrisTW, BieriT, CabunocA, ChanJ, et al. (2011) WormBase 2012: more genomes, more data, new website. Nucleic Acids Res 40 (Database issue): D735–41.

57. HunterS, ApweilerR, AttwoodTK, BairochA, BatemanA, et al. (2009) InterPro: the integrative protein signature database. Nucleic Acids Res 37: D211–215.

58. AbubuckerS, MartinJ, TaylorCM, MitrevaM (2011) HelmCoP: an online resource for helminth functional genomics and drug and vaccine targets prioritization. PLoS One 6: e21832.

59. HillierLW, ReinkeV, GreenP, HirstM, MarraMA, et al. (2009) Massively parallel sequencing of the polyadenylated transcriptome of C. elegans. Genome Res 19: 657–666.

60. Chatr-aryamontriA, CeolA, PalazziLM, NardelliG, SchneiderMV, et al. (2007) MINT: the Molecular INTeraction database. Nucleic Acids Research 35: D572–D574.

61. SmootME, OnoK, RuscheinskiJ, WangPL, IdekerT (2011) Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics 27: 431–432.

62. WarsowG, GreberB, FalkSS, HarderC, SiatkowskiM, et al. (2010) ExprEssence–revealing the essence of differential experimental data in the context of an interaction/regulation net-work. BMC Syst Biol 4: 164.

63. O'BoyleNM, BanckM, JamesCA, MorleyC, VandermeerschT, et al. (2011) Open Babel: An open chemical toolbox. J Cheminform 3: 33.

64. RaoR, WeilGJ (2002) In vitro effects of antibiotics on Brugia malayi worm survival and reproduction. J Parasitol 88: 605–611.

65. EddySR (1998) Profile hidden Markov models. Bioinformatics 14: 755–763.

66. LiBW, RushAC, JiangDJ, MitrevaM, AbubuckerS, et al. (2011) Gender-associated genes in filarial nematodes are important for reproduction and potential intervention targets. PLoS Negl Trop Dis 5: e947.

67. DeLano LW (2006) PyMOL, version 1.2r1. Available: http://www.pymol.org. Accessed 25 September 2007.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 2
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#