#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Utilizing Chemical Genomics to Identify Cytochrome as a Novel Drug Target for Chagas Disease


Chagas Disease, or American trypanosomiasis, is caused by the kinetoplastid protozoan Trypanosoma cruzi and is primarily transmitted to a mammalian host via a triatomine insect vector (the “kissing bug”) infected with T. cruzi parasites. Although discovered in 1909 by the physician Dr. Carlos Chagas, the disease gained recognition by the public health community only following a major outbreak in Brazil during the 1960s. Approximately eight million people (primarily in Central and South America) are infected with T. cruzi and cases are becoming more widespread due to migration out of the endemic regions. Current treatment options have severe problems with toxicity, limited efficacy, and long administration. Hence, discovery of new drugs for treatment of Chagas disease has become of prime interest to the biomedical research community. In this study, we report identification of a potent inhibitor of T. cruzi growth and use a chemical genetics-based approach to elucidate the associated mechanism of action. We found that this compound, GNF7686, targets cytochrome b, a component of the mitochondrial electron transport chain crucial for ATP generation. Our study provides new insights into the use of phenotypic screening to identify novel targets for kinetoplastid drug discovery.


Vyšlo v časopise: Utilizing Chemical Genomics to Identify Cytochrome as a Novel Drug Target for Chagas Disease. PLoS Pathog 11(7): e32767. doi:10.1371/journal.ppat.1005058
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005058

Souhrn

Chagas Disease, or American trypanosomiasis, is caused by the kinetoplastid protozoan Trypanosoma cruzi and is primarily transmitted to a mammalian host via a triatomine insect vector (the “kissing bug”) infected with T. cruzi parasites. Although discovered in 1909 by the physician Dr. Carlos Chagas, the disease gained recognition by the public health community only following a major outbreak in Brazil during the 1960s. Approximately eight million people (primarily in Central and South America) are infected with T. cruzi and cases are becoming more widespread due to migration out of the endemic regions. Current treatment options have severe problems with toxicity, limited efficacy, and long administration. Hence, discovery of new drugs for treatment of Chagas disease has become of prime interest to the biomedical research community. In this study, we report identification of a potent inhibitor of T. cruzi growth and use a chemical genetics-based approach to elucidate the associated mechanism of action. We found that this compound, GNF7686, targets cytochrome b, a component of the mitochondrial electron transport chain crucial for ATP generation. Our study provides new insights into the use of phenotypic screening to identify novel targets for kinetoplastid drug discovery.


Zdroje

1. Anis Rassi Jr AR, Jose Antonio Marin-Neto (2010) Chagas disease. Lancet 375: 1388–1402. doi: 10.1016/S0140-6736(10)60061-X 20399979

2. Magdaleno A, Suarez Mantilla B, Rocha SC, Pral EM, Silber AM (2011) The Involvement of Glutamate Metabolism in the Resistance to Thermal, Nutritional, and Oxidative Stress in Trypanosoma cruzi. Enzyme Res 2011: 486928. doi: 10.4061/2011/486928 21629861

3. Rodolfo Viotti CV, Bruno Lococo, Maria Gabriela Alvarez, Marcos Petti, Graciela Bertocchi and Alejandro Armenti (2009) Side effects of benznidazole as treatment in chronic Chagas disease: fears and realities. Expert Rev Anti Infect Ther 7: 157–163. doi: 10.1586/14787210.7.2.157 19254164

4. Bern C, Kjos S, Yabsley MJ, Montgomery SP (2011) Trypanosoma cruzi and Chagas' Disease in the United States. Clin Microbiol Rev 24: 655–681. doi: 10.1128/CMR.00005-11 21976603

5. Hurwitz I, Fieck A, Klein N, Jose C, Kang A, et al. (2012) A Paratransgenic Strategy for the Control of Chagas Disease. Psyche: A Journal of Entomology 2012: 1–10.

6. Buckner FS, Navabi N (2010) Advances in Chagas disease drug development: 2009–2010. Curr Opin Infect Dis 23: 609–616. doi: 10.1097/QCO.0b013e3283402956 20885320

7. Choi JY, Calvet CM, Gunatilleke SS, Ruiz C, Cameron MD, et al. (2013) Rational development of 4-aminopyridyl-based inhibitors targeting Trypanosoma cruzi CYP51 as anti-chagas agents. J Med Chem 56: 7651–7668. doi: 10.1021/jm401067s 24079662

8. Clayton J (2010) Chagas disease: pushing through the pipeline. Nature 465: S12–15. doi: 10.1038/nature09224 20571548

9. Friggeri L, Hargrove TY, Rachakonda G, Williams AD, Wawrzak Z, et al. (2014) Structural Basis for Rational Design of Inhibitors Targeting Trypanosoma cruzi Sterol 14alpha-Demethylase: Two Regions of the Enzyme Molecule Potentiate its Inhibition. J Med Chem.

10. Bustamante JM, Craft JM, Crowe BD, Ketchie SA, Tarleton RL (2014) New, combined, and reduced dosing treatment protocols cure Trypanosoma cruzi infection in mice. J Infect Dis 209: 150–162. doi: 10.1093/infdis/jit420 23945371

11. Pinazo MJ, Espinosa G, Gallego M, Lopez-Chejade PL, Urbina JA, et al. (2010) Successful treatment with posaconazole of a patient with chronic Chagas disease and systemic lupus erythematosus. Am J Trop Med Hyg 82: 583–587. doi: 10.4269/ajtmh.2010.09-0620 20348503

12. DNDi Press Release. Drug Trial for Leading Parasitic Killer of the Americas Shows Mixed Results but Provides New Evidence for Improved Therapy. Drugs for Neglected Diseases Initiative. 14 November 2013. http://www.dndi.org/media-centre/press-releases/1700-e1224.html. Accessed 01 December 2014.

13. VandeBerg JL (2014) Treatment Trials and Efficacy Determination in Non-human Primates with Chronic T. cruzi Infections. Southwest National Primate Research Center at the Texas Biomedical Research Institute, San Antonio, Texas, USA. pp. 23.

14. Urbina JA (2015) Recent clinical trials for the etiological treatment of chronic chagas disease: advances, challenges and perspectives. J Eukaryot Microbiol 62: 149–156. doi: 10.1111/jeu.12184 25284065

15. McKerrow JH, Doyle PS, Engel JC, Podust LM, Robertson SA, et al. (2009) Two approaches to discovering and developing new drugs for Chagas disease. Mem Inst Oswaldo Cruz 104 Suppl 1: 263–269. 19753483

16. Choy JW, Bryant C, Calvet CM, Doyle PS, Gunatilleke SS, et al. (2013) Chemical-biological characterization of a cruzain inhibitor reveals a second target and a mammalian off-target. Beilstein J Org Chem 9: 15–25. doi: 10.3762/bjoc.9.3 23400640

17. Engel JC, Doyle PS, Hsieh I, McKerrow JH (1998) Cysteine protease inhibitors cure an experimental Trypanosoma cruzi infection. J Exp Med 188: 725–734. 9705954

18. Doyle PS, Zhou YM, Engel JC, McKerrow JH (2007) A cysteine protease inhibitor cures Chagas' disease in an immunodeficient-mouse model of infection. Antimicrob Agents Chemother 51: 3932–3939. 17698625

19. Carneiro G, Aguiar MG, Fernandes AP, Ferreira LA (2012) Drug delivery systems for the topical treatment of cutaneous leishmaniasis. Expert Opin Drug Deliv 9: 1083–1097. doi: 10.1517/17425247.2012.701204 22724539

20. Gilbert IH, Leroy D, Frearson JA (2011) Finding new hits in neglected disease projects: target or phenotypic based screening? Curr Top Med Chem 11: 1284–1291. 21401505

21. Bourguignon SC, de Souza W, Souto-Padron T (1998) Localization of lectin-binding sites on the surface of Trypanosoma cruzi grown in chemically defined conditions. Histochem Cell Biol 110: 527–534. 9826132

22. Garcia-Silva MR, Frugier M, Tosar JP, Correa-Dominguez A, Ronalte-Alves L, et al. (2010) A population of tRNA-derived small RNAs is actively produced in Trypanosoma cruzi and recruited to specific cytoplasmic granules. Mol Biochem Parasitol 171: 64–73. doi: 10.1016/j.molbiopara.2010.02.003 20156490

23. Carpenter AE, Jones TR, Lamprecht MR, Clarke C, Kang IH, et al. (2006) CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol 7: R100. 17076895

24. Engel JC, Ang KK, Chen S, Arkin MR, McKerrow JH, et al. (2010) Image-based high-throughput drug screening targeting the intracellular stage of Trypanosoma cruzi, the agent of Chagas' disease. Antimicrob Agents Chemother 54: 3326–3334. doi: 10.1128/AAC.01777-09 20547819

25. Ke H, Morrisey JM, Ganesan SM, Painter HJ, Mather MW, et al. (2011) Variation among Plasmodium falciparum strains in their reliance on mitochondrial electron transport chain function. Eukaryot Cell 10: 1053–1061. doi: 10.1128/EC.05049-11 21685321

26. Nam TG, McNamara CW, Bopp S, Dharia NV, Meister S, et al. (2011) A chemical genomic analysis of decoquinate, a Plasmodium falciparum cytochrome b inhibitor. ACS Chem Biol 6: 1214–1222. doi: 10.1021/cb200105d 21866942

27. Painter HJ, Morrisey JM, Mather MW, Vaidya AB (2007) Specific role of mitochondrial electron transport in blood-stage Plasmodium falciparum. Nature 446: 88–91. 17330044

28. Plouffe D, Brinker A, McNamara C, Henson K, Kato N, et al. (2008) In silico activity profiling reveals the mechanism of action of antimalarials discovered in a high-throughput screen. Proc Natl Acad Sci U S A 105: 9059–9064. doi: 10.1073/pnas.0802982105 18579783

29. Ghannoum MA, Ibrahim AS, Fu Y, Shafiq MC, Edwards JE Jr., et al. (1992) Susceptibility testing of Cryptococcus neoformans: a microdilution technique. J Clin Microbiol 30: 2881–2886. 1452658

30. Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, et al. (2011) Integrative genomics viewer. Nat Biotechnol 29: 24–26. doi: 10.1038/nbt.1754 21221095

31. Thorvaldsdottir H, Robinson JT, Mesirov JP (2013) Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief Bioinform 14: 178–192. doi: 10.1093/bib/bbs017 22517427

32. Telford JE, Kilbride SM, Davey GP (2010) Decylubiquinone increases mitochondrial function in synaptosomes. J Biol Chem 285: 8639–8645. doi: 10.1074/jbc.M109.079780 20080966

33. Spinazzi M, Casarin A, Pertegato V, Salviati L, Angelini C (2012) Assessment of mitochondrial respiratory chain enzymatic activities on tissues and cultured cells. Nat Protoc 7: 1235–1246. doi: 10.1038/nprot.2012.058 22653162

34. International Glossina Genome I (2014) Genome sequence of the tsetse fly (Glossina morsitans): vector of African trypanosomiasis. Science 344: 380–386. doi: 10.1126/science.1249656 24763584

35. Holmes R K, Jobling M.G. (1996) Medical Microbiology. 4th Edition. Galveston, TX: University of Texas Medical Branch at Galveston,.

36. Lang GI, Rice DP, Hickman MJ, Sodergren E, Weinstock GM, et al. (2013) Pervasive genetic hitchhiking and clonal interference in forty evolving yeast populations. Nature 500: 571–574. doi: 10.1038/nature12344 23873039

37. Lukes J, Guilbride DL, Votypka J, Zikova A, Benne R, et al. (2002) Kinetoplast DNA network: evolution of an improbable structure. Eukaryot Cell 1: 495–502. 12455998

38. Messenger LA, Llewellyn MS, Bhattacharyya T, Franzen O, Lewis MD, et al. (2012) Multiple mitochondrial introgression events and heteroplasmy in trypanosoma cruzi revealed by maxicircle MLST and next generation sequencing. PLoS Negl Trop Dis 6: e1584. doi: 10.1371/journal.pntd.0001584 22506081

39. Giaever G, Shoemaker DD, Jones TW, Liang H, Winzeler EA, et al. (1999) Genomic profiling of drug sensitivities via induced haploinsufficiency. Nat Genet 21: 278–283. 10080179

40. Smith AM, Ammar R, Nislow C, Giaever G (2010) A survey of yeast genomic assays for drug and target discovery. Pharmacol Ther 127: 156–164. doi: 10.1016/j.pharmthera.2010.04.012 20546776

41. Nyfeler B, Hoepfner D, Palestrant D, Kirby CA, Whitehead L, et al. (2012) Identification of elongation factor G as the conserved cellular target of argyrin B. PLoS One 7: e42657. doi: 10.1371/journal.pone.0042657 22970117

42. Richie DL, Thompson KV, Studer C, Prindle VC, Aust T, et al. (2013) Identification and evaluation of novel acetolactate synthase inhibitors as antifungal agents. Antimicrob Agents Chemother 57: 2272–2280. doi: 10.1128/AAC.01809-12 23478965

43. Giaever G, Flaherty P, Kumm J, Proctor M, Nislow C, et al. (2004) Chemogenomic profiling: identifying the functional interactions of small molecules in yeast. Proc Natl Acad Sci U S A 101: 793–798. 14718668

44. Cherry JM HE, Amundsen C, Balakrishnan R, Binkley G, Chan ET, Christie KR, Costanzo MC, Dwight SS, Engel SR, Fisk DG, Hirschman JE, Hitz BC, Karra K, Krieger CJ, Miyasato SR, Nash RS, Park J, Skrzypek MS, Simison M, Weng S, Wong ED (2012) Saccharomyces Genome Database: the genomics resource of budding yeast. Nucleic Acids Res. pp. D700–705.

45. Fisher N, Meunier B (2005) Re-examination of inhibitor resistance conferred by Qo-site mutations in cytochrome b using yeast as a model system. Pest Manag Sci 61: 973–978. 15912560

46. Gutierrez-Cirlos EB, Merbitz-Zahradnik T, Trumpower BL (2004) Inhibition of the yeast cytochrome bc1 complex by ilicicolin H, a novel inhibitor that acts at the Qn site of the bc1 complex. J Biol Chem 279: 8708–8714. 14670947

47. di Rago JP, Coppee JY, Colson AM (1989) Molecular basis for resistance to myxothiazol, mucidin (strobilurin A), and stigmatellin. Cytochrome b inhibitors acting at the center o of the mitochondrial ubiquinol-cytochrome c reductase in Saccharomyces cerevisiae. J Biol Chem 264: 14543–14548. 2547800

48. Trumpower BL (1990) Cytochrome bc1 Complexes of Microorganisms. MICROBIOLOGICAL REVIEWS 54: 101–129. 2163487

49. Huang LS, Cobessi D, Tung EY, Berry EA (2005) Binding of the respiratory chain inhibitor antimycin to the mitochondrial bc1 complex: a new crystal structure reveals an altered intramolecular hydrogen-bonding pattern. J Mol Biol 351: 573–597. 16024040

50. Fisher N, Meunier B (2008) Molecular basis of resistance to cytochrome bc1 inhibitors. FEMS Yeast Res 8: 183–192. 18093133

51. Soni MP, Shelkar N, Gaikwad RV, Vanage GR, Samad A, et al. (2014) Buparvaquone loaded solid lipid nanoparticles for targeted delivery in theleriosis. J Pharm Bioallied Sci 6: 22–30. doi: 10.4103/0975-7406.124309 24459400

52. Peloso Ede F, Vitor SC, Ribeiro LH, Pineyro MD, Robello C, et al. (2011) Role of Trypanosoma cruzi peroxiredoxins in mitochondrial bioenergetics. J Bioenerg Biomembr 43: 419–424. doi: 10.1007/s10863-011-9365-4 21732175

53. Silva TM, Peloso EF, Vitor SC, Ribeiro LH, Gadelha FR (2011) O2 consumption rates along the growth curve: new insights into Trypanosoma cruzi mitochondrial respiratory chain. J Bioenerg Biomembr 43: 409–417. doi: 10.1007/s10863-011-9369-0 21732174

54. Docampo R, Moreno SN, Vercesi AE (1993) Effect of thapsigargin on calcium homeostasis in Trypanosoma cruzi trypomastigotes and epimastigotes. Mol Biochem Parasitol 59: 305–313. 8341327

55. Martins RM, Covarrubias C, Rojas RG, Silber AM, Yoshida N (2009) Use of L-proline and ATP production by Trypanosoma cruzi metacyclic forms as requirements for host cell invasion. Infect Immun 77: 3023–3032. doi: 10.1128/IAI.00138-09 19433547

56. El-Sayed NM, Myler PJ, Blandin G, Berriman M, Crabtree J, et al. (2005) Comparative genomics of trypanosomatid parasitic protozoa. Science 309: 404–409. 16020724

57. Ding MG, di Rago JP, Trumpower BL (2006) Investigating the Qn site of the cytochrome bc1 complex in Saccharomyces cerevisiae with mutants resistant to ilicicolin H, a novel Qn site inhibitor. J Biol Chem 281: 36036–36043. 16987808

58. di Rago J.-P. JP, and Colson A.-M. (1990) Isolation and RNA sequence analysis of cytochrome b mutants resistant to funiculosin, a center i inhibitor of the mitochondrial ubiquinol-cytochrome c reductase in Saccharomyces cerevisiae. FEBS Letters 263: 93–98. 2158909

59. Stringer JR, Beard CB, Miller RF, Wakefield AE (2002) A new name (Pneumocystis jiroveci) for Pneumocystis from humans. Emerg Infect Dis 8: 891–896. 12194762

60. Perocchi F, Jensen LJ, Gagneur J, Ahting U, von Mering C, et al. (2006) Assessing systems properties of yeast mitochondria through an interaction map of the organelle. PLoS Genet 2: e170. 17054397

61. (DNDi) DfNDI (2013) Drug Trial for Leading Parasitic Killer of the Americas Shows Mixed Results but Provides New Evidence for Improved Therapy. Media Center/Press Releases.

62. Molina I, Gomez i Prat J, Salvador F, Trevino B, Sulleiro E, et al. (2014) Randomized trial of posaconazole and benznidazole for chronic Chagas' disease. N Engl J Med 370: 1899–1908. doi: 10.1056/NEJMoa1313122 24827034

63. Andriani G, Amata E, Beatty J, Clements Z, Coffey BJ, et al. (2013) Antitrypanosomal lead discovery: identification of a ligand-efficient inhibitor of Trypanosoma cruzi CYP51 and parasite growth. J Med Chem 56: 2556–2567. doi: 10.1021/jm400012e 23448316

64. Calvet CM, Vieira DF, Choi JY, Kellar D, Cameron MD, et al. (2014) 4-Aminopyridyl-based CYP51 inhibitors as anti-Trypanosoma cruzi drug leads with improved pharmacokinetic profile and in vivo potency. J Med Chem 57: 6989–7005. doi: 10.1021/jm500448u 25101801

65. Diaz-Gonzalez R, Kuhlmann FM, Galan-Rodriguez C, Madeira da Silva L, Saldivia M, et al. (2011) The susceptibility of trypanosomatid pathogens to PI3/mTOR kinase inhibitors affords a new opportunity for drug repurposing. PLoS Negl Trop Dis 5: e1297. doi: 10.1371/journal.pntd.0001297 21886855

66. Hoepfner D, McNamara CW, Lim CS, Studer C, Riedl R, et al. (2012) Selective and specific inhibition of the plasmodium falciparum lysyl-tRNA synthetase by the fungal secondary metabolite cladosporin. Cell Host Microbe 11: 654–663. doi: 10.1016/j.chom.2012.04.015 22704625

67. Rao SP, Lakshminarayana SB, Kondreddi RR, Herve M, Camacho LR, et al. (2013) Indolcarboxamide is a preclinical candidate for treating multidrug-resistant tuberculosis. Sci Transl Med 5: 214ra168. doi: 10.1126/scitranslmed.3007355 24307692

68. Xia D, Yu CA, Kim H, Xia JZ, Kachurin AM, et al. (1997) Crystal structure of the cytochrome bc1 complex from bovine heart mitochondria. Science 277: 60–66. 9204897

69. Kleinschroth T, Castellani M, Trinh CH, Morgner N, Brutschy B, et al. (2011) X-ray structure of the dimeric cytochrome bc(1) complex from the soil bacterium Paracoccus denitrificans at 2.7-A resolution. Biochim Biophys Acta 1807: 1606–1615. doi: 10.1016/j.bbabio.2011.09.017 21996020

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 7
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#