#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Discovery of Anthelmintic Drug Targets and Drugs Using Chokepoints in Nematode Metabolic Pathways


Parasitic roundworm infections plague more than 2 billion people (1/3 of humanity) and cause drastic losses in crops and livestock. New anthelmintic drugs are urgently needed as new drug resistance and environmental concerns arise. A “chokepoint reaction” is defined as a reaction that either consumes a unique substrate or produces a unique product. A chokepoint analysis provides a systematic method of identifying novel potential drug targets. Chokepoint enzymes were identified in the genomes of 10 nematode species, and the intersection and union of all chokepoint enzymes were found. By studying and experimentally testing available compounds known to target proteins orthologous to nematode chokepoint proteins in public databases, this study uncovers features of chokepoints that make them successful drug targets. Chemogenomic screening was performed on drug-like compounds from public drug databases to find existing compounds that target homologs of nematode chokepoints. The compounds were prioritized based on chemical properties frequently found in successful drugs and were experimentally tested using Caenorhabditis elegans. Several drugs that are already known anthelmintic drugs and novel candidate targets were identified. Seven of the compounds were tested in Caenorhabditis elegans and three yielded a detrimental phenotype. One of these three drug-like compounds, Perhexiline, also yielded a deleterious effect in Haemonchus contortus and Onchocerca lienalis, two nematodes with divergent forms of parasitism. Perhexiline, known to affect the fatty acid oxidation pathway in mammals, caused a reduction in oxygen consumption rates in C. elegans and genome-wide gene expression profiles provided an additional confirmation of its mode of action. Computational modeling of Perhexiline and its target provided structural insights regarding its binding mode and specificity. Our lists of prioritized drug targets and drug-like compounds have potential to expedite the discovery of new anthelmintic drugs with broad-spectrum efficacy.


Vyšlo v časopise: Discovery of Anthelmintic Drug Targets and Drugs Using Chokepoints in Nematode Metabolic Pathways. PLoS Pathog 9(8): e32767. doi:10.1371/journal.ppat.1003505
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003505

Souhrn

Parasitic roundworm infections plague more than 2 billion people (1/3 of humanity) and cause drastic losses in crops and livestock. New anthelmintic drugs are urgently needed as new drug resistance and environmental concerns arise. A “chokepoint reaction” is defined as a reaction that either consumes a unique substrate or produces a unique product. A chokepoint analysis provides a systematic method of identifying novel potential drug targets. Chokepoint enzymes were identified in the genomes of 10 nematode species, and the intersection and union of all chokepoint enzymes were found. By studying and experimentally testing available compounds known to target proteins orthologous to nematode chokepoint proteins in public databases, this study uncovers features of chokepoints that make them successful drug targets. Chemogenomic screening was performed on drug-like compounds from public drug databases to find existing compounds that target homologs of nematode chokepoints. The compounds were prioritized based on chemical properties frequently found in successful drugs and were experimentally tested using Caenorhabditis elegans. Several drugs that are already known anthelmintic drugs and novel candidate targets were identified. Seven of the compounds were tested in Caenorhabditis elegans and three yielded a detrimental phenotype. One of these three drug-like compounds, Perhexiline, also yielded a deleterious effect in Haemonchus contortus and Onchocerca lienalis, two nematodes with divergent forms of parasitism. Perhexiline, known to affect the fatty acid oxidation pathway in mammals, caused a reduction in oxygen consumption rates in C. elegans and genome-wide gene expression profiles provided an additional confirmation of its mode of action. Computational modeling of Perhexiline and its target provided structural insights regarding its binding mode and specificity. Our lists of prioritized drug targets and drug-like compounds have potential to expedite the discovery of new anthelmintic drugs with broad-spectrum efficacy.


Zdroje

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

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

3. Osei-AtweneboanaMY, EngJK, BoakyeDA, GyapongJO, PrichardRK (2007) Prevalence and intensity of Onchocerca volvulus infection and efficacy of ivermectin in endemic communities in Ghana: a two-phase epidemiological study. Lancet 369: 2021–2029.

4. GeertsS, GryseelsB (2000) Drug resistance in human helminths: current situation and lessons from livestock. Clin Microbiol Rev 13: 207–222.

5. WeilGJ, KastensW, SusapuM, LaneySJ, WilliamsSA, et al. (2008) The impact of repeated rounds of mass drug administration with diethylcarbamazine plus albendazole on bancroftian filariasis in Papua New Guinea. PLoS Negl Trop Dis 2: e344.

6. BarkerKR, HusseyRS, KrusbergLR, BirdGW, DunnRA, et al. (1994) Plant and soil nematodes: societal impact and focus for the future. J Nematol 26: 127–137.

7. ZasadaIA, HalbrendtJM, Kokalis-BurelleN, LamondiaJ, McKenryMV, et al. (2010) Managing Nematodes Without Methyl Bromide. Annu Rev Phytopathol 48: 311–28.

8. YehI, HanekampT, TsokaS, KarpPD, AltmanRB (2004) Computational analysis of Plasmodium falciparum metabolism: organizing genomic information to facilitate drug discovery. Genome Res 14: 917–924.

9. PalumboMC, ColosimoA, GiulianiA, FarinaL (2007) Essentiality is an emergent property of metabolic network wiring. FEBS Lett 581: 2485–2489.

10. SinghS, MalikBK, SharmaDK (2007) Choke point analysis of metabolic pathways in E.histolytica: a computational approach for drug target identification. Bioinformation 2: 68–72.

11. PerumalD, LimCS, SakharkarKR, SakharkarMK (2009) ‘Load Points’ and ‘Choke Points’ as Nodes for Prioritizing Drug Targets in Pseudomonas aeruginosa. Current Bioinformatics 4: 48–53.

12. RahmanSA, SchomburgD (2006) Observing local and global properties of metabolic pathways: ‘load points’ and ‘choke points’ in the metabolic networks. Bioinformatics 22: 1767–1774.

13. FatumoS, PlaimasK, MallmJP, SchrammG, AdebiyiE, et al. (2009) Estimating novel potential drug targets of Plasmodium falciparum by analysing the metabolic network of knock-out strains in silico. Infect Genet Evol 9: 351–358.

14. KimTY, KimHU, LeeSY (2010) Metabolite-centric approaches for the discovery of antibacterials using genome-scale metabolic networks. Metab Eng 12: 105–111.

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

16. MitrevaM, ZarlengaDS, McCarterJP, JasmerDP (2007) Parasitic nematodes - from genomes to control. Vet Parasitol 148: 31–42.

17. BlaxterML, De LeyP, GareyJR, LiuLX, ScheldemanP, et al. (1998) A molecular evolutionary framework for the phylum Nematoda. Nature 392: 71–75.

18. KanehisaM, GotoS, FurumichiM, TanabeM, HirakawaM (2010) KEGG for representation and analysis of molecular networks involving diseases and drugs. Nucleic Acids Res 38: D355–360.

19. WishartDS, KnoxC, GuoAC, ChengD, ShrivastavaS, et al. (2008) DrugBank: a knowledgebase for drugs, drug actions and drug targets. Nucleic Acids Res 36: D901–906.

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

21. OppermanCH, BirdDM, WilliamsonVM, RokhsarDS, BurkeM, et al. (2008) Sequence and genetic map of Meloidogyne hapla: A compact nematode genome for plant parasitism. Proc Natl Acad Sci U S A 105: 14802–14807.

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

23. DieterichC, CliftonSW, SchusterLN, ChinwallaA, DelehauntyK, et al. (2008) The Pristionchus pacificus genome provides a unique perspective on nematode lifestyle and parasitism. Nat Genet 40: 1193–1198.

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

25. BrownleeDJ, Holden-DyeL, WalkerRJ (1997) Actions of the anthelmintic ivermectin on the pharyngeal muscle of the parasitic nematode, Ascaris suum. Parasitology 115 ((Pt 5)) 553–561.

26. JasmerDP, YaoC, RehmanA, JohnsonS (2000) Multiple lethal effects induced by a benzimidazole anthelmintic in the anterior intestine of the nematode Haemonchus contortus. Mol Biochem Parasitol 105: 81–90.

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

28. ShannonP, MarkielA, OzierO, BaligaNS, WangJT, et al. (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res 13: 2498–2504.

29. LipinskiCA, LombardoF, DominyBW, FeeneyPJ (2001) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 46: 3–26.

30. HuangSC, FreitasTC, AmielE, EvertsB, PearceEL, et al. (2012) Fatty acid oxidation is essential for egg production by the parasitic flatworm Schistosoma mansoni. PLoS Pathog 8: e1002996.

31. MorgulisA, GertzEM, SchafferAA, AgarwalaR (2006) A fast and symmetric DUST implementation to mask low-complexity DNA sequences. Journal of computational biology : a journal of computational molecular cell biology 13: 1028–1040.

32. TrapnellC, PachterL, SalzbergSL (2009) TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25: 1105–1111.

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

34. RobinsonMD, McCarthyDJ, SmythGK (2010) edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26: 139–140.

35. ZdobnovEM, ApweilerR (2001) InterProScan–an integration platform for the signature-recognition methods in InterPro. Bioinformatics 17: 847–848.

36. GoujonM, McWilliamH, LiW, ValentinF, SquizzatoS, et al. (2010) A new bioinformatics analysis tools framework at EMBL-EBI. Nucleic Acids Res 38: W695–699.

37. AshburnerM, BallCA, BlakeJA, BotsteinD, ButlerH, et al. (2000) Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25: 25–29.

38. BenjaminiY, HochbergY (1995) Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. Journal of the Royal Statistical Society Series B (Methodological) 57: 289–300.

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

40. SchiothHB, FredrikssonR (2005) The GRAFS classification system of G-protein coupled receptors in comparative perspective. General and comparative endocrinology 142: 94–101.

41. MorrisGM, HueyR, LindstromW, SannerMF, BelewRK, et al. (2009) AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J Comput Chem 30: 2785–2791.

42. HsiaoYS, JoglG, EsserV, TongL (2006) Crystal structure of rat carnitine palmitoyltransferase II (CPT-II). Biochem Biophys Res Commun 346: 974–980.

43. 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.

44. McGarryJD, BrownNF (1997) The mitochondrial carnitine palmitoyltransferase system. From concept to molecular analysis. Eur J Biochem 244: 1–14.

45. ZubovychIO, StraudS, RothMG (2010) Mitochondrial dysfunction confers resistance to multiple drugs in Caenorhabditis elegans. Mol Biol Cell 21: 956–969.

46. RamsayRR, ZammitVA (2004) Carnitine acyltransferases and their influence on CoA pools in health and disease. Mol Aspects Med 25: 475–493.

47. ParkEA, CookGA (1998) Differential regulation in the heart of mitochondrial carnitine palmitoyltransferase-I muscle and liver isoforms. Mol Cell Biochem 180: 27–32.

48. MorrisGM, HueyR, LindstromW, SannerMF, BelewRK, et al. (2009) AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of computational chemistry 30: 2785–2791.

49. NoroesJ, DreyerG, SantosA, MendesVG, MedeirosZ, et al. (1997) Assessment of the efficacy of diethylcarbamazine on adult Wuchereria bancrofti in vivo. Trans R Soc Trop Med Hyg 91: 78–81.

50. DoumboO, RossignolJF, PichardE, TraoreHA, DembeleTM, et al. (1997) Nitazoxanide in the treatment of cryptosporidial diarrhea and other intestinal parasitic infections associated with acquired immunodeficiency syndrome in tropical Africa. Am J Trop Med Hyg 56: 637–639.

51. CampbellWC, FisherMH, StapleyEO, Albers-SchonbergG, JacobTA (1983) Ivermectin: a potent new antiparasitic agent. Science 221: 823–828.

52. MartinRJ, RobertsonAP, BjornH (1997) Target sites of anthelmintics. Parasitology 114 Suppl: S111–124.

53. PrichardRK (1973) The fumarate reductase reaction of Haemonchus contortus and the mode of action of some anthelmintics. Int J Parasitol 3: 409–417.

54. PrichardRK (1970) Mode of action of the anthelminthic thiabendazole in Haemonchus contortus. Nature 228: 684–685.

55. BerrimanM, HaasBJ, LoVerdePT, WilsonRA, DillonGP, et al. (2009) The genome of the blood fluke Schistosoma mansoni. Nature 460: 352–358.

56. GiordaniRB, WeizenmannM, RosembergDB, De CarliGA, BogoMR, et al. (2010) Trichomonas vaginalis nucleoside triphosphate diphosphohydrolase and ecto-5′-nucleotidase activities are inhibited by lycorine and candimine. Parasitol Int 59: 226–231.

57. SansomFM, RobsonSC, HartlandEL (2008) Possible effects of microbial ecto-nucleoside triphosphate diphosphohydrolases on host-pathogen interactions. Microbiol Mol Biol Rev 72: 765–781, Table of Contents.

58. GounarisK, ThomasS, NajarroP, SelkirkME (2001) Secreted variant of nucleoside diphosphate kinase from the intracellular parasitic nematode Trichinella spiralis. Infect Immun 69: 3658–3662.

59. GhoshI, RaghavanN, FitzGeraldPC, ScottAL (1995) Nucleoside diphosphate kinase from the parasitic nematode Brugia malayi. Gene 164: 261–266.

60. SajidMS, IqbalZ, MuhammadG, IqbalMU (2006) Immunomodulatory effect of various anti-parasitics: a review. Parasitology 132: 301–313.

61. HarrisTW, AntoshechkinI, BieriT, BlasiarD, ChanJ, et al. (2010) WormBase: a comprehensive resource for nematode research. Nucleic Acids Res 38: D463–467.

62. TanakaM, GilbertJ, PappanoAJ (1992) Inhibition of sodium pump by l-palmitoylcarnitine in single guinea-pig ventricular myocytes. J Mol Cell Cardiol 24: 711–719.

63. HarunaT, HorieM, TakanoM, KonoY, YoshidaH, et al. (2000) Alteration of the membrane lipid environment by L-palmitoylcarnitine modulates K(ATP) channels in guinea-pig ventricular myocytes. Pflugers Arch 441: 200–207.

64. KennedyJA, UngerSA, HorowitzJD (1996) Inhibition of carnitine palmitoyltransferase-1 in rat heart and liver by perhexiline and amiodarone. Biochem Pharmacol 52: 273–280.

65. CattoBA (1981) Schistosoma mansoni: decarboxylation of 5-hydroxytryptophan, L-dopa, and L-histidine in adult and larval schistosomes. Exp Parasitol 51: 152–157.

66. BoutrosM, AhringerJ (2008) The art and design of genetic screens: RNA interference. Nat Rev Genet 9: 554–566.

67. QuW, RenC, LiY, ShiJ, ZhangJ, et al. (2011) Reliability analysis of the Ahringer Caenorhabditis elegans RNAi feeding library: a guide for genome-wide screens. BMC Genomics 12: 170.

68. SimmerF, MoormanC, van der LindenAM, KuijkE, van den BerghePV, et al. (2003) Genome-wide RNAi of C. elegans using the hypersensitive rrf-3 strain reveals novel gene functions. PLoS Biol 1: E12.

69. ChanM, TanDS, SimTS (2007) Plasmodium falciparum pyruvate kinase as a novel target for antimalarial drug-screening. Travel Med Infect Dis 5: 125–131.

70. ZoraghiR, SeeRH, Axerio-CiliesP, KumarNS, GongH, et al. (2011) Identification of pyruvate kinase in methicillin-resistant Staphylococcus aureus as a novel antimicrobial drug target. Antimicrob Agents Chemother 55: 2042–2053.

71. van PoeljePD, PotterSC, ErionMD (2011) Fructose-1, 6-bisphosphatase inhibitors for reducing excessive endogenous glucose production in type 2 diabetes. Handb Exp Pharmacol 279–301.

72. MelnikovA, ZaborinaO, DhimanN, PrabhakarBS, ChakrabartyAM, et al. (2000) Clinical and environmental isolates of Burkholderia cepacia exhibit differential cytotoxicity towards macrophages and mast cells. Mol Microbiol 36: 1481–1493.

73. YuasaK, Mi-IchiF, KobayashiT, YamanouchiM, KoteraJ, et al. (2005) PfPDE1, a novel cGMP-specific phosphodiesterase from the human malaria parasite Plasmodium falciparum. Biochem J 392: 221–229.

74. ShakurY, de KoningHP, KeH, KambayashiJ, SeebeckT (2011) Therapeutic potential of phosphodiesterase inhibitors in parasitic diseases. Handb Exp Pharmacol 487–510.

75. MennitiFS, FaraciWS, SchmidtCJ (2006) Phosphodiesterases in the CNS: targets for drug development. Nat Rev Drug Discov 5: 660–670.

76. SanonA, Tournaire-ArellanoC, El HageSY, BoriesC, CaujolleR, et al. (2005) N-acetyl-beta-D-hexosaminidase from Trichomonas vaginalis: substrate specificity and activity of inhibitors. Biomed Pharmacother 59: 245–248.

77. VillelaAD, Sanchez-QuitianZA, DucatiRG, SantosDS, BassoLA (2011) Pyrimidine salvage pathway in Mycobacterium tuberculosis. Curr Med Chem 18: 1286–1298.

78. KucerovaL, MatuskovaM, PastorakovaA, TyciakovaS, JakubikovaJ, et al. (2008) Cytosine deaminase expressing human mesenchymal stem cells mediated tumour regression in melanoma bearing mice. J Gene Med 10: 1071–1082.

79. LinCH, ChungMY, ChenWB, ChienCH (2007) Growth inhibitory effect of the human NIT2 gene and its allelic imbalance in cancers. FEBS J 274: 2946–2956.

80. BatemanRL, BhanumoorthyP, WitteJF, McClardRW, GrompeM, et al. (2001) Mechanistic inferences from the crystal structure of fumarylacetoacetate hydrolase with a bound phosphorus-based inhibitor. J Biol Chem 276: 15284–15291.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 8
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#