#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Real-Time Imaging of the Intracellular Glutathione Redox Potential in the Malaria Parasite


In the malaria parasite Plasmodium falciparum, the cellular redox potential influences signaling events, antioxidant defense, and mechanisms of drug action and resistance. Until now, the real-time determination of the redox potential in malaria parasites has been limited because conventional approaches disrupt sub-cellular integrity. Using a glutathione biosensor comprising human glutaredoxin-1 linked to a redox-sensitive green fluorescent protein (hGrx1-roGFP2), we systematically characterized basal values and drug-induced changes in the cytosolic glutathione-dependent redox potential (EGSH) of drug-sensitive (3D7) and resistant (Dd2) P. falciparum parasites. Via confocal microscopy, we demonstrated that hGrx1-roGFP2 rapidly detects EGSH changes induced by oxidative and nitrosative stress. The cytosolic basal EGSH of 3D7 and Dd2 were estimated to be −314.2±3.1 mV and −313.9±3.4 mV, respectively, which is indicative of a highly reducing compartment. We furthermore monitored short-, medium-, and long-term changes in EGSH after incubation with various redox-active compounds and antimalarial drugs. Interestingly, the redox cyclers methylene blue and pyocyanin rapidly changed the fluorescence ratio of hGrx1-roGFP2 in the cytosol of P. falciparum, which can, however, partially be explained by a direct interaction with the probe. In contrast, quinoline and artemisinin-based antimalarial drugs showed strong effects on the parasites' EGSH after longer incubation times (24 h). As tested for various conditions, these effects were accompanied by a drop in total glutathione concentrations determined in parallel with alternative methods. Notably, the effects were generally more pronounced in the chloroquine-sensitive 3D7 strain than in the resistant Dd2 strain. Based on these results hGrx1-roGFP2 can be recommended as a reliable and specific biosensor for real-time spatiotemporal monitoring of the intracellular EGSH in P. falciparum. Applying this technique in further studies will enhance our understanding of redox regulation and mechanisms of drug action and resistance in Plasmodium and might also stimulate redox research in other pathogens.


Vyšlo v časopise: Real-Time Imaging of the Intracellular Glutathione Redox Potential in the Malaria Parasite. PLoS Pathog 9(12): e32767. doi:10.1371/journal.ppat.1003782
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003782

Souhrn

In the malaria parasite Plasmodium falciparum, the cellular redox potential influences signaling events, antioxidant defense, and mechanisms of drug action and resistance. Until now, the real-time determination of the redox potential in malaria parasites has been limited because conventional approaches disrupt sub-cellular integrity. Using a glutathione biosensor comprising human glutaredoxin-1 linked to a redox-sensitive green fluorescent protein (hGrx1-roGFP2), we systematically characterized basal values and drug-induced changes in the cytosolic glutathione-dependent redox potential (EGSH) of drug-sensitive (3D7) and resistant (Dd2) P. falciparum parasites. Via confocal microscopy, we demonstrated that hGrx1-roGFP2 rapidly detects EGSH changes induced by oxidative and nitrosative stress. The cytosolic basal EGSH of 3D7 and Dd2 were estimated to be −314.2±3.1 mV and −313.9±3.4 mV, respectively, which is indicative of a highly reducing compartment. We furthermore monitored short-, medium-, and long-term changes in EGSH after incubation with various redox-active compounds and antimalarial drugs. Interestingly, the redox cyclers methylene blue and pyocyanin rapidly changed the fluorescence ratio of hGrx1-roGFP2 in the cytosol of P. falciparum, which can, however, partially be explained by a direct interaction with the probe. In contrast, quinoline and artemisinin-based antimalarial drugs showed strong effects on the parasites' EGSH after longer incubation times (24 h). As tested for various conditions, these effects were accompanied by a drop in total glutathione concentrations determined in parallel with alternative methods. Notably, the effects were generally more pronounced in the chloroquine-sensitive 3D7 strain than in the resistant Dd2 strain. Based on these results hGrx1-roGFP2 can be recommended as a reliable and specific biosensor for real-time spatiotemporal monitoring of the intracellular EGSH in P. falciparum. Applying this technique in further studies will enhance our understanding of redox regulation and mechanisms of drug action and resistance in Plasmodium and might also stimulate redox research in other pathogens.


Zdroje

1. MurrayCJ, RosenfeldLC, LimSS, AndrewsKG, ForemanKJ, et al. (2012) Global malaria mortality between 1980 and 2010: a systematic analysis. Lancet 379: 413–431.

2. EastmanRT, FidockDA (2009) Artemisinin-based combination therapies: A vital tool in efforts to eliminate malaria. Nat Rev Microbiol 7: 864–874.

3. Krauth-SiegelRL, BauerH, SchirmerRH (2005) Dithiol proteins as guardians of the intracellular redox milieu in parasites: old and new drug targets in trypanosomes and malaria-causing plasmodia. Angew Chem Int Ed Engl 44: 690–715.

4. RahlfsS, BeckerK (2006) Interference with redox-active enzymes as a basis for the design of antimalarial drugs. Mini Rev Med Chem 6: 163–76.

5. KehrS, SturmN, RahlfsS, PrzyborskiJM, BeckerK (2010) Compartmentation of redox metabolism in malaria parasites. PLoS Pathog 23: 6(12).

6. FidockDA, RosenthalPJ, CroftSL, BrunR, NwakaS (2004) Antimalarial drug discovery: efficacy models for compound screening. Nat Rev Drug Discov 3: 509–20.

7. BeckerK, TilleyL, VennerstromJL, RobertsD, RogersonS, et al. (2004) Oxidative stress in malaria parasite-infected erythrocytes: host-parasite interactions. Int J Parasitol 34: 163–89.

8. Komaki-YasudaK, KawazuS, KanoS (2003) Disruption of the Plasmodium falciparum 2-Cys peroxiredoxin gene renders parasites hypersensitive to reactive oxygen and nitrogen species. FEBS Lett 547: 140–4.

9. WangDY, WuYL (2000) A possible antimalarial action mode of qinghaosu (artemisinin) series compounds. Alkylation of reduced glutathione by C-centered primary radicals produced from antimalarial compound qinghaosu and 12-(2,4-dimethoxyphenyl)-12-deoxoqinghaosu. Chem Commun 5: 2193–2194.

10. O'NeillPM, PosnerGH (2004) A medicinal chemistry perspective on artemisinin and related endoperoxides. J Med Chem 47: 2945–64.

11. FoleyM, TilleyL (1998) Quinoline antimalarials: mechanisms of action and resistance and prospects for new agents. Pharmacol Ther 79: 55–87.

12. AtamnaH, KrugliakM, ShalmievG, DeharoE, PescarmonaG, et al. (1996) Mode of antimalarial effect of methylene blue and some of its analogues on Plasmodium falciparum in culture and their inhibition of P. vinckei petteri and P. yoelii nigeriensis in vivo. Biochemical Pharmacology 51: 693–700.

13. FaminO, KrugliakM, GinsburgH (1999) Kinetics of inhibition of glutathione-mediated degradation of ferriprotoporphyrin IX by antimalarial drugs. Biochem Pharmacol 58: 59–68.

14. LoriaP, MillerS, FoleyM, TilleyL (1999) Inhibition of the peroxidative degradation of haem as the basis of action of chloroquine and other quinoline antimalarials. Biochem J 339: 363–370.

15. GravesPR, KwiekJJ, FaddenP, RayR, HardemanK, et al. (2002) Discovery of novel targets of quinoline drugs in the human purine binding proteome. Mol Pharmacol 62: 1364–1372.

16. MontiD, BasilicoN, ParapiniS, PasiniE, OlliaroP, et al. (2002) Does chloroquine really act through oxidative stress? Fed Eur Biochem Soc Lett 522: 3–5.

17. FärberPM, ArscottLD, WilliamsCH, BeckerK, SchirmerRH (1998) Recombinant Plasmodium falciparum glutathione reductase is inhibited by the antimalarial dye methylene blue. FEBS Lett 422: 311–314.

18. Pastrana-MenaR, DinglasanRR, Franke-FayardB, Vega-RodríguezJ, Fuentes-CaraballoM, et al. (2010) Glutathione reductase-null malaria parasites have normal blood stage growth but arrest during development in the mosquito. J Biol Chem 285: 27045–56.

19. BuchholzK, PutriantiED, RahlfsS, SchirmerRH, BeckerK, et al. (2010) Molecular genetics evidence for the in vivo roles of the two major NADPH-dependent disulfide reductases in the malaria parasite. J Biol Chem 285: 37388–95.

20. PatzewitzEM, Salcedo-SoraJE, WongEH, SethiaS, StocksPA, et al. (2013) Glutathione transport: A new role for PfCRT in chloroquine resistance. Antioxid Redox Signal 19: 683–685.

21. MeierjohannS, WalterRD, MullerS (2002) Regulation of intracellular glutathione levels in erythrocytes infected with chloroquine-sensitive and chloroquine-resistant Plasmodium falciparum. Biochem J 368: 761–768.

22. BeckerK, RahlfsS, NickelC, SchirmerRH (2003) Glutathione - function and metabolism in the malarial parasite Plasmodium falciparum. Biol Chem 348: 551–566.

23. JortzikE, BeckerK (2012) Thioredoxin and glutathione systems in Plasmodium falciparum. Int J Med Microbiol 302: 187–94.

24. SchaferFQ, BuettnerGR (2001) Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic Biol Med 30: 1191–1212.

25. GreveB, LehmanLG, LellB, LucknerD, Schmidt-OttR, et al. (1999) High oxygen radical production is associated with fast parasite clearance in children with Plasmodium falciparum malaria. J Infect Dis 179: 1584–1586.

26. BozdechZ, GinsburgH (2004) Antioxidant defense in Plasmodium falciparum-data mining of the transcriptome. Malaria Journal 3: 23.

27. AtamnaH, GinsburgH (1997) The malaria parasite supplies glutathione to its host cell investigation of glutathione transport and metabolism in human erythrocytes infected with Plasmodium falciparum. Eur J Biochem 250: 670–679.

28. MorganB, SobottaMC, DickTP (2011) Measuring E(GSH) and H2O2 with roGFP2-based redox probes. Free Radic Biol Med 51: 1943–51.

29. MandavilliBS, JanesMS (2010) Detection of intracellular glutathione using ThiolTracker Violet stain and fluorescence microscopy. Curr Protoc Cytom. Chapter 9: Unit 9.35.

30. DardalhonM, KumarC, IraquiI, VernisL, KiendaG, et al. (2012) Redox-sensitive YFP sensors monitor dynamic nuclear and cytosolic glutathione redox changes. Free Radic Biol Med 52: 2254–2265.

31. DooleyCT, DoreTM, HansonGT, JacksonWC, RemingtonSJ, et al. (2004) Imaging dynamic redox changes in mammalian cells with green fluorescent protein indicators. J Biol Chem 279: 22284–93.

32. GutscherM, PauleauAL, MartyL, BrachT, WabnitzGH, et al. (2008) Real-time imaging of the intracellular glutathione redox potential. Nat Methods 5: 553–559.

33. MeyerAJ, BrachT, MartyL, KreyeS, RouhierN, et al. (2007) Redox-sensitive GFP in Arabidopsis thaliana is a quantitative biosensor for the redox potential of the cellular glutathione redox buffer. Plant J 52: 973–86.

34. CrabbBS, RugM, GilbergerTW, ThompsonJK, TrigliaT, et al. (2004) Transfection of the human malaria parasite Plasmodium falciparum. Methods Mol Biol 270: 263–276.

35. SchwarzländerM, FrickerMD, MüllerC, MartyL, BrachT, et al. (2008) Confocal imaging of glutathione redox potential in living plant cells. J Microsc 231: 299–316.

36. KasoziDM, GromerS, AdlerH, ZocherK, RahlfsS, et al. (2011) The bacterial redox signaller pyocyanin as an anti-plasmodial agent: comparisons with its thioanalog methylene blue. Redox Rep 16: 154–65.

37. BauerH, Fritz-WolfK, WinzerA, KühnerS, LittleS, et al. (2006) A fluoro analogue of the menadione derivative 6-[2′-(3′-methyl)-1′, 4′-naphthoquinolyl]hexanoic acid is a suicide substrate of glutathione reductase. Crystal structure of the alkylated human enzyme. J Am Chem Soc 128: 10784–94.

38. AkoachereM, BuchholzK, FischerE, BurhenneJ, HaefeliWE, et al. (2005) In vitro assessment of methylene blue on chloroquine-sensitive and -resistant Plasmodium falciparum strains reveals synergistic action with artemisinins. Antimicrob Agents Chemother 49: 4592–4597.

39. WhiteNJ (2008) Qinghaosu (artemisinin): the price of success. Science 320: 330–4.

40. RajDK, MuJ, JiangH, KabatJ, SinghS, et al. (2009) Disruption of a Plasmodium falciparum multidrug resistance-associated protein (PfMRP) alters its fitness and transport of antimalarial drugs and glutathione. J Biol Chem 284: 7687–96.

41. LuersenK, WalterRD, MullerS (2000) Plasmodium falciparum infected red blood cells depend on a functional glutathione de novo synthesis attributable to an enhanced loss of glutathione. Biochem J 346: 545–552.

42. KehrS, JortzikE, DelahuntyC, YatesJR3rd, RahlfsS, et al. (2011) Protein S-glutathionylation in malaria parasites. Antioxid Redox Signal 15: 2855–65.

43. KuhnY, RohrbachP, LanzerM (2007) Quantitative pH measurements in Plasmodium falciparum-infected erythrocytes using pHluorin. Cell Microbiol 9: 1004–13.

44. SalibaKJ, KirkK (1999) pH regulation in the intracellular malaria parasite, Plasmodium falciparum. H(+) extrusion via a v-type H(+)-ATPase. J Biol Chem 274: 33213–33219.

45. HayashiM, YamadaH, MitamuraT, HoriiT, YamamotoA, et al. (2000) Vacuolar H(+)-ATPase localized in plasma membranes of malaria parasite cells, Plasmodium falciparum, is involved in regional acidification of parasitized erythrocytes. J Biol Chem 275: 34353–34358.

46. BraunNA, MorganB, DickTP, SchwappachB (2010) The yeast CLC protein counteracts vesicular acidification during iron starvation. J Cell Sci 123: 2342–2350.

47. GuttmannP, EhrlichP (1891) über die Wirkung des Methylenblau bei Malaria. BerlinKlin Wochenschr 28: 953–956.

48. CoulibalyB, ZoungranaA, MockenhauptFP, SchirmerRH, KloseC, et al. (2009) Strong gametocytocidal effect of methylene blue-based combination therapy against falciparum malaria: a randomised controlled trial. PLoS One 4: e5318.

49. SchirmerRH, AdlerH, PickhardtM, MandelkowE (2011) “Lest we forget you-methylene blue…”. Neurobiol Aging 32: 232–2325.

50. ZoungranaA, CoulibalyB, SiéA, Walter-SackI, MockenhauptFP, et al. (2008) Safety and efficacy of methylene blue combined with artesunate or amodiaquine for uncomplicated falciparum malaria: a randomized controlled trial from Burkina Faso. PLoS One 3: e1630.

51. HartwigCL, RosenthalAS, D'AngeloJ, GriffinCE, PosnerGH, et al. (2009) Accumulation of artemisinin trioxane derivatives within neutral lipids of Plasmodium falciparum malaria parasites is endoperoxide-dependent. Biochem Pharmacol 77: 322–36.

52. del Pilar CrespoM, AveryTD, HanssenE, FoxE, RobinsonTV, et al. (2008) Artemisinin and a series of novel endoperoxide antimalarials exert early effects on digestive vacuole morphology. Antimicrob Agents Chemother 52: 98–109.

53. Eckstein-LudwigU, WebbRJ, Van GoethemID, EastJM, LeeAG, et al. (2003) Artemisinins target the SERCA of Plasmodium falciparum. Nature 424: 957–961.

54. BhisutthibhanJ, PanXQ, HosslerPA, WalkerDJ, YowellCA, et al. (1998) The Plasmodium falciparum translationally controlled tumor protein homolog and its reaction with the antimalarial drug artemisinin. J Biol Chem 273: 16192–16198.

55. KannanR, SahalD, ChauhanVS (2002) Heme-artemisinin adducts are crucial mediators of the ability of artemisinin to inhibit heme polymerization. Chem Biol 9: 321–332.

56. MukanganyamaS, NaikYS, WiderstenM, MannervikB, HaslerJA (2001) Proposed reductive metabolism of artemisinin by glutathione transferases in vitro. Free Radic Res 35: 427–34.

57. LiW, MoW, ShenD, SunL, WangJ, et al. (2005) Yeast model uncovers dual roles of mitochondria in action of artemisinin. PLoS Genet 1: e36.

58. AchanJ, TalisunaAO, ErhartA, YekaA, TibenderanaJK, et al. (2011) Quinine, an old anti-malarial drug in a modern world: role in the treatment of malaria. Malar J 24: 10–144.

59. FidockDA, NomuraT, TalleyAK, CooperRA, DzekunovSM, et al. (2000) Mutations in the P. falciparum digestive vacuole transmembrane protein PfCRT and evidence for their role in chloroquine resistance. Mol Cell 6: 861–871.

60. ReedMB, SalibaKJ, CaruanaSR, KirkK, CowmanAF (2000) Pgh1 modulates sensitivity and resistance to multiple antimalarials in Plasmodium falciparum. Nature 403: 906–909.

61. GinsburgH, FaminO, ZhangJ, KrugliakM (1998) Inhibition of glutathione-dependent degradation of heme by chloroquine and amodiaquine as a possible basis for their antimalarial mode of action. Biochem Pharmacol 56: 1305–1313.

62. WangL, YangZ, FuJ, YinH, XiongK, et al. (2012) Ethaselen: a potent mammalian thioredoxin reductase 1 inhibitor and novel organoselenium anticancer agent. Free Radic Biol Med 52: 898–908.

63. IttaratW, SreepianA, SrisarinA, PathepchotivongK (2003) Effect of dihydroartemisinin on the antioxidant capacity of P. falciparum-infected erythrocytes. Southeast Asian J Trop Med Public Health 34: 744–50.

64. TownsendDM (2007) S-glutathionylation: indicator of cell stress and regulator of the unfolded protein response. Mol Interv 7: 313–24.

65. TragerW, JensenJB (1976) Human malaria parasites in continuous culture. Science 193: 673–675.

66. LambrosC, VanderbergJP (1979) Synchronisation of Plasmodium falciparum erythrocytic stages in culture. J Parasitol 65: 418–420.

67. DesjardinsRE, CanfieldCJ, HaynesJD, ChulayJD (1979) Quantitative assessment of antimalarial activity in vitro by a semiautomated microdilution technique. Antimicrob Agents Chemother 16: 710–718.

68. PrietoH, KoncarevicS, ParkSK, YatesJIII, BeckerK (2008) Large scale differential proteome analysis in Plasmodium falciparum under drug treatment. PLoS One 3: e4098.

69. SanchezCP, DaveA, SteinWD, LanzerM (2010) Transporters as mediators of drug resistance in Plasmodium falciparum. Int J Parasitol 40: 1109–18.

70. SturmN, JortzikE, MailuBM, KoncarevicS, DeponteM, et al. (2009) Identification of proteins targeted by the thioredoxin superfamily in Plasmodium falciparum. PLoS Pathog 5: e1000383.

71. RibautC, BerryA, ChevalleyS, ReybierK, MorlaisI, et al. (2008) Concentration and purification by magnetic separation of the erythrocytic stages of all human Plasmodium species. Malar J 5: 7–45.

72. EllmannGL (1959) Tissue sulfhydryl groups. Arch Biochem Biophys 82: 70–77.

73. BeckerK, GuiM, TraxlerA, KirstenC, SchirmerRH (1994) Redox processes in malaria and other parasitic diseases. Histochemistry 102: 389–395.

74. JiangK, SchwarzerC, LallyE, ZhangS, RuzinS, et al. (2006) Expression and characterization of a redox-sensing green fluorescent protein (reduction-oxidation-sensitive green fluorescent protein) in Arabidopsis. Plant Physiol 141: 397–403.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 12
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#