Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2003 Oct;47(10):3368–3370. doi: 10.1128/AAC.47.10.3368-3370.2003

Activity of Megazol, a Trypanocidal Nitroimidazole, Is Associated with DNA Damage

Bertin Enanga 1, Mark R Ariyanayagam 2, Mhairi L Stewart 1, Michael P Barrett 1,*
PMCID: PMC201145  PMID: 14506061

Abstract

DNA damage associated with the trypanocidal activity of megazol [2-amino-5-(1-methyl-5-nitro-2-imidazolyl)-1,3,4-thiadiazole] was shown in experiments in which DNA repair-deficient RAD51−/− Trypanosoma brucei mutants were found to be hypersensitive to the drug. Parasites resistant to megazol were selected and showed modest cross-resistance to other trypanocides, although neither drug efflux nor changes to intracellular thiols correlated with resistance.


New drugs are urgently required for treatment of human African trypanosomiasis (22). One compound with promise is megazol (5, 12, 13, 18), a nitroheterocyclic compound that forms a nitro radical anion upon reduction (31). Megazol appears to enter Trypanosoma brucei via passive diffusion (3), but little is known about its mode of action. We determined the effect of megazol in assays that measured oxidative and reductive stress. We also selected parasites resistant to megazol in order to assess cellular changes associated with resistance.

Bloodstream forms (17) and procyclic forms (6) of T. brucei brucei (strain 427) were cultivated by using standard techniques. Assays measuring drug sensitivity were carried out to determine the drug concentration producing a 50% decrease in cell proliferation (IC50) and the resistance factor (average ratio of the IC50 for a resistant clone to the IC50 for a wild-type clone). The Alamar Blue test (28) was used for bloodstream forms, while cell counting by using an improved Neubauer chamber was required for procyclics. For procyclic cells the IC50 of megazol was 0.28 ± 0.01 μM, and for bloodstream forms the IC50 was 0.15 ± 0.02 μM.

To induce megazol resistance in vitro, trypanosomes were exposed to drug concentrations that doubled every 2 weeks, starting at 0.008 and 0.01 μM for bloodstream forms and procyclics, respectively. After 6 months of cultivation, procyclic and bloodstream forms that tolerated 10 and 1 μM megazol, respectively, were cloned by limiting dilution.

The IC50 for the cloned, resistant, procyclic line was 29.24 ± 3.2 μM (105-fold-reduced sensitivity), while the IC50 for the bloodstream form line was 3.2 ± 0.48 μM (21-fold-reduced sensitivity).

Cross-resistance to megazol and other trypanocides was also studied (Table 1). Moderate levels of cross-resistance to two nitroheterocyclic trypanocides, the nitrofuran nifurtimox and the nitroimidazole benznidazole, were detected. Significant, albeit low-level, cross-resistance to the melaminophenyl arsenical compounds cymelarsan, melarsen oxide, and melarsoprol, the diamidine compounds pentamidine and berenil, and suramin was also observed.

TABLE 1.

Cross-resistance of bloodstream and procyclic forms of T. brucei brucei strain 427 to megazol and other trypanocidesa

Drug IC50 (μM)
Procyclic forms
Bloodstream forms
Wild type Resistant (factor of resistance) Wild type Resistant (factor of resistance)
Megazol 0.28 ± 0.01 29.24 ± 3.18 (104)* 0.15 ± 0.02 3.20 ± 0.48 (21)*
Nifurtimox 4.89 ± 0.12 37.12 ± 3.84 (7.6)* 5.58 ± 0.86 34.41 ± 4.91 (6.2)*
Benznidazole 2.89 ± 0.20 12.87 ± 1.29 (4.5)** 15.26 ± 3.85 77.36 ± 4.92 (5.2)*
Suramin 15.59 ± 2.61 66.06 ± 4.15 (4.2)** 0.13 ± 0.01 0.32 ± 0.21 (2.5)***
Berenil 10.99 ± 1.50 41.14 ± 1.31 (3.7)* 0.99 ± 0.12 5.18 ± 1.14 (5.2)*
Pentamidine 2.14 ± 0.03 5.06 ± 0.23 (2.4) (NS) 0.04 ± 0.07 0.15 ± 0.01 (3.8)*
Melarsen oxide 0.02 ± 0.00 0.07 ± 0.08 (3.5)* 0.01 ± 0.00 0.05 ± 0.01 (5.0)*
Melarsoprol 2.38 ± 0.08 2.60 ± 0.02 (1.1) (NS) 0.13 ± 0.01 0.30 ± 0.01 (2.3)***
Cymelarsan 0.06 ± 0.00 0.13 ± 0.00 (2.2)*** 0.02 ± 0.02 0.07 ± 0.03 (3.5)***
Ethidium bromide 12.15 ± 1.52 16.89 ± 1.73 (1.4) (NS) 2.07 ± 0.68 4.29 ± 0.71 (2.1) (NS)
Isometamidium 9.80 ± 0.79 11.44 ± 0.79 (1.2) (NS) 3.35 ± 0.56 4.33 ± 0.67 (1.3) (NS)
Brilliant green 0.016 ± 0.00 0.053 ± 00 (3.3) 0.003 ± 00 0.003 ± 0.000 (1.0)
a

Sensitivities of wild-type and selected megazol-resistant lines to a number of drugs were determined. The factors of resistance indicated in parentheses were calculated as ratios of IC50s for resistant parasites to those for wild-type parasites. Values that were significantly different from that for the control, by Student's t test, are designated by an asterisk(s) (*, P < 0.001; **, P < 0.01; ***, P < 0.05; NS, not significant at P = 0.05). Values are means ± standard deviations (n = 3 independent experiments).

Verapamil (20), PAK-104P (7), and two phenothiazine derivatives (prochlorperazine and trifluoperazine) (14) all inhibit cellular extrusion pumps but, when used at 5 μM, failed to reverse megazol resistance in T. brucei. Efflux pumps are thus unlikely to play a significant role in the megazol resistance characterized here.

It was recently shown that megazol exposure reduces trypanothione levels in Trypanosoma cruzi (23). Bloodstream form T. brucei obtained from infected Wistar rats (21) and procyclic forms grown in vitro (6) were resuspended at a concentration of 107 cells ml−1 in culture medium. Megazol (0.15 to 30 μM) or buthionine sulfoximine (BSO; 10 to 20 μM) was added to the parasite suspensions, and the suspensions were incubated for 2 h. Reduced thiol levels in control and drug-treated parasites were then determined by derivatization with monobromobimane (thiolyte) and separation by high-pressure liquid chromatography as previously described (30).

BSO (16) reduces glutathione (2) and trypanothione (15) levels in trypanosomatids. When added to cell cultures 24 h prior to evaluation of IC50s, BSO enhanced the susceptibilities of both wild-type and megazol-resistant trypanosomes to megazol. At 10 and 20 μM, respectively, BSO caused the IC50 for wild-type procyclics to drop from 0.28 to 0.14 ± 0.02 μM and that for resistant cells to drop from 29.24 to 7.33 ± 0.56 μM. With bloodstream forms, the IC50 for wild-type cells shifted from 0.15 to 0.07 ± 0.01 μM while that for resistant cells shifted from 3.2 to 1.03 ± 0.03 μM. The concentration of BSO could not exceed 20 μM since BSO is highly toxic to T. brucei (IC50s were 15.15 ± 0.22 μM and 11 ± 0.39 μM for procyclic and bloodstream forms, respectively), although related parasitic trypanosomatids T. cruzi (26) and Leishmania (19) can tolerate doses of BSO of 1 mM. The BSO effect was greater in resistant than in wild-type T. brucei.

The effects of BSO and megazol (used at their in vitro IC50s) on the combined thiol levels (total levels of glutathione, trypanothione, and glutathionylspermidine and, measurable only in procyclics [1], of ovothiol) in wild-type and resistant trypanosomes of both forms were determined. Megazol treatment, for 2 h, did not have a significant impact on thiol content in bloodstream or procyclic forms, while BSO, at the concentrations used here, affected only the thiol content of procyclic forms (reducing it about twofold from 3.2 nmol [108 cells]−1).

No significant difference in total thiol levels was observed between wild-type and resistant forms, indicating that alterations to thiol levels are not involved in the development of resistance characterized here.

Nitroheterocyclic compounds are generally believed to exert their cytotoxic effects only after activation by single electron reduction of their corresponding nitro anion radicals (27). In bacteria, the toxic effects of metronidazole are accompanied by damage to DNA (25, 29) and cells defective in DNA repair become hypersensitive to the action of metronidazole (11). A principal enzyme involved in eukaryotic DNA repair is RAD51. T. brucei mutants lacking this enzyme (24) were hypersensitive to megazol, indicating that the toxic effects of megazol are accompanied by damage to DNA (Table 2). In contrast, the susceptibility of RAD51 deletion mutants to the trypanocidal nitrofuran nifurtimox was similar to that of wild-type cells. N-acetylcysteine (NAC) antagonizes oxidative stress by interacting with a number of the reduced oxygen species (4, 8). When coadministered at 0.5 mM (the highest concentration at which no detrimental effect on trypanosomes was induced by this compound), NAC had no antagonistic effect on the action of megazol while it did have a modest protective effect against the action of nifurtimox (Table 2).

TABLE 2.

Activity of megazol against wild-type and RAD51−/− T. brucei bloodstream formsa

Drug IC50 (μM)
Wild type RAD51−/− mutants
Megazol 0.13 ± 0.0025 0.029 ± 0.002
Megazol with NAC 0.13 ± 0.0020 ND
Nifurtimox 3.37 ± 0.19 3.40 ± 0.15
Nifurtimox with NAC 12.72 ± 1.1 ND
a

Values are means ± standard deviations (n = 3 independent experiments). The effect of 0.5 mM NAC was assessed by adding this concentration to wild-type cells only prior to addition of drug. ND, not determined.

The activity of nifurtimox in T. brucei therefore appears to be mediated through reduced oxygen species, produced during futile redox cycling with reduced drug. Megazol's activity, however, derives from direct damage exerted by the reduced megazol nitro anion radical derivatives. In T. cruzi, nifurtimox is also known to exert its activity through redox cycling (9, 10). Recently it was shown that nifurtimox stimulated an increase in oxygen uptake into T. cruzi cells while megazol did not (23). Thus, experiments using different approaches and different species of trypanosomes indicate that the activities of the nitrofuran nifurtimox and the nitroimidazole megazol arise through different mechanisms.

Acknowledgments

Bertin Enanga was a Wellcome Trust Traveling Fellow.

The excellent assistance of Flora Logan (funded by a Society for General Microbiology summer studentship) and Janice Brock is gratefully acknowledged. Richard McCulloch (University of Glasgow) kindly donated the RAD51−/− strain of T. brucei.

REFERENCES

  • 1.Ariyanayagam, M. R., and A. H. Fairlamb. 2001. Ovothiol and trypanothione as antioxidants in trypanosomatids. Mol. Biochem. Parasitol. 115:189-198. [DOI] [PubMed] [Google Scholar]
  • 2.Arrick, B. A., O. W. Griffith., and A. Cerami. 1981. Inhibition of glutathione synthesis as a chemotherapeutic strategy for trypanosomiasis. J. Exp. Med. 153:720-725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Barrett, M. P., A. H. Fairlamb, B. Rousseau, G. Chauviere, and J. Perie. 2000. Uptake of the nitroimidazole drug megazol by African trypanosomes. Biochem. Pharmacol. 59:615-620. [DOI] [PubMed] [Google Scholar]
  • 4.Benrahmoune, M., P. Therond, and Z. Abedinzadeh. 2000. The reaction of superoxide radical with N-acetylcysteine. Free Radic. Biol. Med. 29:775-782. [DOI] [PubMed] [Google Scholar]
  • 5.Bouteille, B., A. Marie-Daragon, G. Chauviere, C. de Albuquerque, B Enanga, M. L. Darde, J. M. Vallat, J. Perie, and M. Dumas. 1995. Effect of megazol on Trypanosoma brucei brucei acute and subacute infections in Swiss mice. Acta Trop. 60:73-80. [DOI] [PubMed] [Google Scholar]
  • 6.Brun, R., and M. Schonenberger. 1979. Cultivation and in vitro cloning of procyclic culture forms of Trypanosoma brucei in a semi-defined medium. Acta Trop. 36:289-292. [PubMed] [Google Scholar]
  • 7.Chuman, Y., Z. S. Chen, K. Seto, T. Sumizawa, T. Furukawa, A. Tani, M. Haraguchi, K. Niwa, K. Yamada, T. Aikou, and S. Akiyama. 1998. Reversal of MRP-mediated vincristine resistance in KB cells by buthionine sulfoximine in combination with PAK-104P. Cancer Lett. 129:69-76. [DOI] [PubMed] [Google Scholar]
  • 8.Cotgreave, I. A. 1997. N-acetylcysteine: pharmacological considerations and experimental and clinical applications. Adv. Pharmacol. 38:205-227. [PubMed] [Google Scholar]
  • 9.Docampo, R., and A. O. Stoppani. 1979. Generation of superoxide anion and hydrogen peroxide induced by nifurtimox in Trypanosoma cruzi. Arch. Biochem. Biophys. 197:317-321. [DOI] [PubMed] [Google Scholar]
  • 10.Docampo, R., S. N. Moreno, A. O. Stoppani, W. Leon, F. S. Cruz, F. Villalta, and R. F. Muniz. 1981. Mechanism of nifurtimox toxicity in different forms of Trypanosoma cruzi. Biochem. Pharmacol. 30:1947-1951. [DOI] [PubMed] [Google Scholar]
  • 11.Elizondo, G., M. E. Gonsebatt, A. M. Salazar, I. Lares, P. Santiago, J. Herrera, E. Hong, and P. Ostrosky-Wegman. 1996. Genotoxic effects of metronidazole. Mutat. Res. 370:75-80. [DOI] [PubMed] [Google Scholar]
  • 12.Enanga, B., M. Keita, G. Chauviere, M. Dumas, and B. Bouteille. 1998. Megazol combined with suramin: a chemotherapy regimen which reversed the CNS pathology in a model of human African trypanosomiasis in mice. Trop. Med. Int. Health 3:736-741. [DOI] [PubMed] [Google Scholar]
  • 13.Enanga, B., J. M. Ndon, H. Boudra, L. Debrauwer, G. Dubreuil, B. Bouteille, G. Chauviere, C. Labat, M. Dumas, J. Perie, and G. Houin. 2000. Pharmacokinetics, metabolism and excretion of megazol in a Trypanosoma brucei gambiense primate model of human African trypanosomiasis. Preliminary study. Arzneim.-Forsch. 50:158-162. [DOI] [PubMed] [Google Scholar]
  • 14.Essodaigui, M., F. Frezard, E. S. Moreira, F. Dagger, and A. Garnier-Suillerot. 1999. Energy-dependent efflux from Leishmania promastigotes of substrates of the mammalian multidrug resistance pumps. Mol. Biochem. Parasitol. 100:73-84. [DOI] [PubMed] [Google Scholar]
  • 15.Fairlamb, A. H., and A. Cerami. 1992. Metabolism and functions of trypanothione in the kinetoplastida. Annu. Rev. Microbiol. 46:695-729. [DOI] [PubMed] [Google Scholar]
  • 16.Griffith, O. W., and A. Meister. 1979. Potent and specific inhibition of glutathione synthesis by buthionine sulfoximine (S-n-butyl homocysteine sulfoximine). J. Biol. Chem. 254:7558-7560. [PubMed] [Google Scholar]
  • 17.Hirumi, H., and K. Hirumi. 1989. Continuous cultivation of Trypanosoma brucei brucei bloodstream forms in a medium containing a low concentration of serum protein without feeder cell layers. J. Parasitol. 75:985-989. [PubMed] [Google Scholar]
  • 18.Jennings, F. W., G. Chauviere, C. Viode, and M. Murray. 1996. Topical chemotherapy for experimental African trypanosomiasis with cerebral involvement: the use of melarsoprol combined with the 5-nitroimidazole, megazol. Trop. Med. Int. Health 1:363-366. [DOI] [PubMed] [Google Scholar]
  • 19.Kapoor, P., M. Sachdev, and R. Madhubala. 2000. Inhibition of glutathione synthesis as a chemotherapeutic strategy for leishmaniasis. Trop. Med. Int. Health 5:438-442. [DOI] [PubMed] [Google Scholar]
  • 20.Kaur, J., and C. S. Dey. 2000. Putative P-glycoprotein expression in arsenite-resistant Leishmania donovani down-regulated by verapamil. Biochem. Biophys. Res. Commun. 271:615-619. [DOI] [PubMed] [Google Scholar]
  • 21.Lanham, S. M., and D. G. Godfrey. 1970. Isolation of salivarian trypanosomes from man and other mammals using DEAE-cellulose. Exp. Parasitol. 28:521-534. [DOI] [PubMed] [Google Scholar]
  • 22.Legros, D., G. Ollivier, M. Gastellu-Etchegorry, C. Paquet, C. Burri, J. Jannin, and P. Buscher. 2002. Treatment of human African trypanosomiasis—present situation and needs for research and development. Lancet Infect. Dis. 2:437-440. [DOI] [PubMed] [Google Scholar]
  • 23.Maya, J. D., S. Bollo, L. J. Nunez-Vergara, J. A. Squella, Y. Repetto, A. Morello, J. Perie, and G. Chauviere. 2003. Trypanosoma cruzi: effect and mode of action of nitroimidazole and nitrofuran derivatives. Biochem. Pharmacol. 65:999-1006. [DOI] [PubMed] [Google Scholar]
  • 24.McCulloch, R., and J. D. Barry. 1999. A role for RAD51 and homologous recombination in Trypanosoma brucei antigenic variation. Genes Dev. 13:2875-2888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Mendz, G. L., and F. Megraud. 2002. Is the molecular basis of metronidazole resistance in microaerophilic organisms understood? Trends Microbiol. 10:370-375. [DOI] [PubMed] [Google Scholar]
  • 26.Mocanda, C., Y. Repetto, J. Aldunate, M. E. Letelier, and A. Morello. 1989. Role of glutathione in the susceptibility of Trypanosoma cruzi to drugs. Comp. Biochem. Physiol. 94C:87-91. [DOI] [PubMed]
  • 27.Moreno, S. N., and R. Docampo. 1985. Mechanism of toxicity of nitro compounds used in the chemotherapy of trichomoniasis. Environ. Health Perspect. 64:199-208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Raz, B., M. Iten, Y. Grether-Buhler, R. Kaminsky, and R. Brun. 1997. The Alamar Blue assay to determine drug sensitivity of African trypanosomes (T. b. rhodesiense and T. b. gambiense) in vitro. Acta Trop. 68:139-147. [DOI] [PubMed] [Google Scholar]
  • 29.Sander, P., K. G. Papavinasasundaram, T. Dick, E. Stavropoulos, K. Ellrott, B. Springer, M. J. Colston, and E. C. Böttger. 2001. Mycobacterium bovis BCG recA deletion mutant shows increased susceptibility to DNA-damaging agents but wild-type survival in a mouse infection model. Infect. Immun. 69:3562-3568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shim, H., and A. H. Fairlamb. 1988. Levels of polyamines, glutathione and glutathione-spermidine conjugates during growth of the insect trypanosomatid Crithidia fasciculata. J. Gen. Microbiol. 134:807-817. [DOI] [PubMed] [Google Scholar]
  • 31.Viode, C., N. Bettach, N. Cenas, R. L. Krauth-Siegel, G. Chauviere, N. Bakalara, and J. Perie. 1999. Enzymatic reduction studies of nitroheterocycles. Biochem. Pharmacol. 57:549-557. [DOI] [PubMed] [Google Scholar]

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES