Next Article in Journal
Predicting Anticancer Drug Resistance Mediated by Mutations
Next Article in Special Issue
Pyrvinium Pamoate and Structural Analogs Are Early Macrofilaricide Leads
Previous Article in Journal
Tunicamycin Protects against LPS-Induced Lung Injury
Previous Article in Special Issue
Discovery, Development, Inventions and Patent Review of Fexinidazole: The First All-Oral Therapy for Human African Trypanosomiasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Laboratory Selection of Trypanosomatid Pathogens for Drug Resistance

by
Sabina Beilstein
1,2,
Radhia El Phil
3,4,
Suzanne Sherihan Sahraoui
3,4,
Leonardo Scapozza
3,4,
Marcel Kaiser
1,2 and
Pascal Mäser
1,2,*
1
Department Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
2
Swiss TPH, University of Basel, Petersplatz 1, 4002 Basel, Switzerland
3
School of Pharmaceutical Sciences, University of Geneva, 1205 Geneva, Switzerland
4
Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(2), 135; https://doi.org/10.3390/ph15020135
Submission received: 23 December 2021 / Revised: 13 January 2022 / Accepted: 14 January 2022 / Published: 24 January 2022
(This article belongs to the Collection Drug Discovery and Development for Tropical Diseases (TDs))

Abstract

:
The selection of parasites for drug resistance in the laboratory is an approach frequently used to investigate the mode of drug action, estimate the risk of emergence of drug resistance, or develop molecular markers for drug resistance. Here, we focused on the How rather than the Why of laboratory selection, discussing different experimental set-ups based on research examples with Trypanosoma brucei, Trypanosoma cruzi, and Leishmania spp. The trypanosomatids are particularly well-suited to illustrate different strategies of selecting for drug resistance, since it was with African trypanosomes that Paul Ehrlich performed such an experiment for the first time, more than a century ago. While breakthroughs in reverse genetics and genome editing have greatly facilitated the identification and validation of candidate resistance mutations in the trypanosomatids, the forward selection of drug-resistant mutants still relies on standard in vivo models and in vitro culture systems. Critical questions are: is selection for drug resistance performed in vivo or in vitro? With the mammalian or with the insect stages of the parasites? Under steady pressure or by sudden shock? Is a mutagen used? While there is no bona fide best approach, we think that a methodical consideration of these questions provides a helpful framework for selection of parasites for drug resistance in the laboratory.

1. The TriTryp Parasites

Trypanosoma brucei, Trypanosoma cruzi and Leishmania comprise the human-pathogenic species in the trypanosomatid family. They cause the neglected tropical diseases sleeping sickness, Chagas disease and leishmaniasis, which have an estimated total prevalence of over 10 million and impose a substantial burden on global health [1,2]. The insect vectors—tsetse flies, triatomine bugs and phlebotomine sandflies, respectively—transmit the parasites to the mammalian host during a blood meal. The parasites thus encounter very different environments in their transmission cycles. Furthermore, the three species are zoonotic and infect various mammals. Leishmania and T. cruzi are intracellular in the mammalian host and extracellular in the gut of the insect, whereas all life-cycle stages of T. brucei are extracellular. This renders T. brucei easier to cultivate than T. cruzi or Leishmania. In addition, T. brucei is more readily amenable to reverse genetics than other trypanosomatids [3]. Table 1 compares the cellular and molecular characteristics of the TriTryp parasites T. brucei, T. cruzi and L. donovani. The drugs for treating the respective diseases are listed in Table 2, along with the in vitro sensitivity of the different life-cycle stages of the parasites. These drugs were developed by phenotypic, cell-based approaches rather than target-based. Consequently, for some of them, even though they have been used for decades, the mechanism of action is still not fully understood.

2. New Tools for Target Deconvolution

Knowing a drug’s target is of great importance for the development of more effective, better tolerated therapies and for the management of drug resistance. However, the genetic mapping of mutations conferring resistance, which in Caenorhabditis elegans was key to the identification of anthelmintic drug targets [15], is precluded in trypanosomatids as sexual recombination, if it occurs at all, is not obligate. This lack of forward genetics is to some extent compensated for by the declining costs of next-generation DNA sequencing, which have made it affordable to identify resistance mutations by whole genome sequencing or transcriptome sequencing of drug-resistant mutants [16,17]. In addition, reverse genetic tools were developed that have helped to overcome the lack of forward genetics.
Inducible RNA interference (RNAi) libraries were used as a high-throughput method for genome-wide loss-of-function studies in T. brucei [18,19]. Performing RNAi induction followed by drug selection confirmed the role of the known drug resistance genes TbAT1, AAT6 and NTR1 as determinants of susceptibility to melarsoprol, eflornithine and nifurtimox, respectively [20,21]. An experimental trypanocide whose target was validated by RNAi is 4-[5-(4-phenoxyphenyl)-2H-pyrazol-3-yl]morpholine, a molecule that turned out to function as a hyperactivator of T. brucei adenosine kinase [22,23]. These approaches obviously require the presence of a functional RNAi system in the target cell (Table 1). This, however, is not the case for T. cruzi and most species of Leishmania, with the notable exception of L. braziliensis [9]. Parsimony suggests that the common trypanosomatid ancestor had been competent of RNAi and that the genes for Argonaute and Dicer proteins were lost multiple times in the subsequent course of evolution [24].
Genome editing by CRISPR-Cas9 is more generally applicable. First established for T. cruzi [12,13,25], it was also successfully applied to Leishmania [14,26] and T. brucei [11,27,28]. Further improvements simplified the genetic manipulation of the trypanosomatids, providing a high-throughput system for large-scale genetic knock-out screens [11,29,30]. Another high-throughput tool that has helped to understand the molecular genetics of drug action in Leishmania are cosmid libraries. Cos-Seq is based on the selection for enriched loci under drug pressure with subsequent sequencing and candidate gene identification [31,32]. Targeted overexpression of candidate genes is possible as well; with DNA repair genes, this has provided new insights into the mode of action of benznidazole in T. cruzi [33]. Inducible gene overexpression libraries have been developed for trypanosomatids that allow identifying drug targets by screening for genes whose overexpression causes drug resistance [34].
Proteomic techniques are applicable to trypanosomatid parasites as well [35,36]. Chemical proteomics combine chemistry to synthesize a drug-derived probe with biology to search for the target protein. This is achieved either by affinity chromatography, in which the chemical probe is immobilized on a matrix and incubated with a cell lysate to fish for proteins, or in situ, where the probe is added to live cells and cross-linked to target proteins e.g., by means of a photoreactive group [37]. The first approach was used to identify MAP kinases and cdc2-related kinases as putative targets of 2,4-diaminopyrimidines in T. brucei [38]. The second identified candidate targets of the antiobesity drug orlistat and the antichagasic protease inhibitor K11777 [39,40]. Other innovative techniques for target fishing were developed, such as protein chips, phage display, or the yeast three-hybrid system [41,42], but have to our knowledge not yet been applied to trypanosomatids.

3. Artificial Selection for Drug Resistance

Selecting pathogens for drug resistance is a classical experiment in a parasitology laboratory. The first scientist known to have performed it was Paul Ehrlich, the father of chemotherapy. Ehrlich and co-workers infected mice with African trypanosomes and treated the animals with subcurative doses of parafuchsin. They observed that after several passages, the trypanosomes had lost their susceptibility to the drug [43]. Decades later, Alexander Fleming observed the same phenomenon when he cultured bacteria on plates with sublethal concentrations of penicillin [44]. Ehrlich’s main interest in drug resistance was to learn about the nature of the subcellular drug targets. He proposed to use artificially selected drug-resistant pathogens as a ‘therapeutic sieve’ based on cross-resistance profiles [45]. Thus, by testing a new drug candidate against a panel of resistant strains, he was able to tell whether it had a different mode of action. A hundred years later, this concept was incorporated for the development of antimalarials [46]. Today, the selection of drug-resistant mutants in the laboratory mainly serves three purposes: (i) to learn about the mode of drug action, i.e., to identify drug transport pathways and drug targets; (ii) to estimate the risk of emergence of drug resistance in the field based on how quickly resistance evolves in the laboratory; and (iii) to find molecular markers for drug resistance that enable rapid, DNA-based tests. In the following, we focus on the How rather than the Why in the experimental process of laboratory-selection for drug resistance, illustrating different protocols with examples from the ‘TriTryp’ parasites.

3.1. Selection In Vitro vs. In Vivo

The selection for drug-resistant trypanosomatids by subcurative dosing of a rodent model of infection is the closest situation to what happens in a treated patient. Therefore, the knowledge that will be gained about the genes and mutations involved in drug resistance is likely to be relevant for the situation in the clinics. A second advantage of selecting in vivo is the high numbers of parasites that can be reached in an infected animal, increasing the probability of success in obtaining a drug-resistant mutant. The main point against in vivo selection is the use of animals per se, if it can be replaced by an in vitro system. In addition, in vivo studies are usually more laborious and expensive than in vitro experiments. A further advantage of in vitro systems is the better control over parameters such as drug concentration and number of parasites.
Overall, there has been a good correlation between the drug resistance phenotypes obtained in vivo and in vitro [47], except in one case, where bloodstream-form T. b. brucei, which had been selected in vivo for Cymelarsan resistance, were only weakly resistant to arsenicals in culture [48,49]. A possible explanation is the fast metabolization of melamine-based arsenicals in vivo [50]. Some mechanisms, such as phenomena involving tissue tropism, will only evolve in vivo, whereas others might occur only in vitro. The latter is exemplified by the finding that expression of a particular variant surface glycoprotein (VSG) in T. brucei causes suramin resistance [51]. Such a mechanism is hardly sustainable in vivo, where the parasites will be eliminated by the adaptive immune response unless they switch to express another VSG.
The first studies on the selection of trypanosomatids for drug resistance were performed in vivo, as long-term culture systems were unavailable at the time. Early studies with T. brucei mainly focused on arsenical resistance [52,53,54,55,56] and on the phenomenon of cross-resistance between melamine-based arsenicals and diamidines [57]. Generally, T. brucei spp. were propagated in immunosuppressed mice. The animals were treated with increasing but subcurative concentrations of arsenicals, and the relapsing trypanosomes were passaged to new mice [48,58,59,60]. In a typical experiment, it took eight rounds of infection to obtain a stable resistance phenotype with a resistance factor of 15 [59]. There are only a few studies where T. cruzi or Leishmania were selected for drug resistance in vivo. Two different approaches were applied, both successfully, to obtain benznidazole-resistant T. cruzi. In the first, selection was performed by repeated treatment shocks: an infected mouse at peak parasitemia was treated with a single oral dose of 500 mg/kg benznidazole and after 6 h, the surviving blood trypomastigotes were inoculated into another mouse. This procedure was repeated about 10 days later, again at the peak of parasitemia. After 25 rounds, the obtained T. cruzi were unresponsive to benznidazole and cross-resistant to nifurtimox and other nitroimidazoles [61]. The second approach used a lower but constant dose: infected mice were given an oral dose of 100 mg/kg benznidazole daily for 20 consecutive days. Thereafter, the animals were immunosuppressed with cyclophosphamide, and the emerging trypomastigotes in the blood were harvested and inoculated into new mice. Four out of five T. cruzi isolates became unresponsive to benznidazole after 2 to 9 rounds of selection [62]. A similar attempt to generate drug-resistant mutants of L. infantum and L. donovani by repeated subcurative dosing of infected hamsters succeeded remarkably quickly for paromomycin, but not for miltefosine [63]. A different approach, pre-exposing animals before infection, was used to test a possible link between the presence of arsenic in the drinking water and the emergence of antimonial resistance in leishmania. Mice that had been given drinking water with 10 ppm arsenite for 1 month were infected with L. donovani. After 28 days, still with arsenite in the animals’ drinking water, the leishmania were passaged to a new group of arsenite pre-exposed mice. After 5 such rounds, the leishmania had become unresponsive to a dose of 500 µg/mL when tested in vitro [64].
There have been many reports on the successful in vitro selection of trypanosomatids for drug resistance (Table 3).
Early in vitro selection experiments were performed even before it was possible to propagate the parasites in culture: African trypanosomes were isolated from an infected rodent at peak parasitemia, exposed in vitro for 1 h to high concentrations of tryparsamide, and reinjected into another animal. After 3 to 13 cycles, the trypanosomes had become less sensitive to tryparsamide [100]. The first culture systems for trypanosomatids were established for the insect stages, i.e., the procyclic, trypomastigote form of T. brucei [101], the epimastigote form of T. cruzi [102], and the promastigote form of Leishmania [103]. When maintained in appropriate medium at 27 °C, these forms readily proliferate in axenic culture and reach densities of over 107 cells per ml before they enter stationary phase. The cultivation of the mammalian stages is less straightforward. Axenic in vitro cultivation is possible for T. brucei bloodstream forms [104]. Amastigote Leishmania, too, can proliferate in axenic culture if they are kept at low pH to simulate the phagolysosome [105]. Axenic long-term cultivation of T. cruzi amastigotes has been reported [106] but is not a standard procedure. Overall, the insect stages of trypanosomatid parasites do not require host cells, are easier to culture than the mammalian stages, and they reach much higher cell densities favoring the selection of drug-resistant mutants. The obvious drawback is that the insect stages are not clinically relevant.

3.2. Selection of Insect Stages vs. Mammalian Stages

The key question regarding the choice of the life cycle-stage is whether the mode of drug action is preserved in the insect stages, including drug transport pathway(s) and intracellular target(s). We would argue that if there is no difference in drug susceptibility between mammalian and insect stages, the mode of drug action is likely to be preserved and hence, in vitro selection for drug resistance will be easier and faster with the insect stages even though it is not the stage causing pathogenesis in mammals. However, if the insect stages are clearly less susceptible than the mammalian stages, they might not provide valuable insights on drug resistance (even though the target could be preserved but not essential in the insect stage). Benznidazole for T. cruzi is a typical example of a drug that is equally active against either stage, epimastigotes and amastigotes [107] (Table 2). Epimastigote T. cruzi selected for benznidazole resistance kept the phenotype after transformation to amastigotes [75]. In contrast, paromomycin resistance that had been selected for in vivo was lost after the amastigote Leishmania had been transformed to promastigotes [63]. Regarding T. brucei, most drugs used for the treatment of sleeping sickness are much less effective against the procyclic forms in the tsetse fly midgut than against the mammalian bloodstream forms (Table 2). This might be due to the fact that some of the transporters mediating drug uptake are only expressed in the latter [108,109].
An interesting aspect about the relevance of selecting insect stages of trypanosomatid parasites for drug resistance is the question of whether the insect stages ever come into contact with drugs in nature. With African trypanosomes, a scenario that seems plausible is that of an infected tsetse fly that takes a blood meal on a cow that has received nagana drugs, whereupon the procyclic trypanosomes in the fly midgut will be exposed to sublethal drug concentrations. This was experimentally reproduced: Glossina morsitans infected with T. congolense were fed over 1 month on rabbits that received weekly prophylactic doses of 2 mg/kg Samorin (isometamidium chloride). The flies were then used to infect mice, which in turn served to infect a new group of teneral flies. After four such cycles, the selected T. congolense had a significantly lower susceptibility to Samorin than unselected ones, passaged in untreated animals [110].

3.3. Selection of a Clone vs. a Population

The probability of obtaining a drug-resistant mutant increases with the genetic diversity of the starting population. This might suggest starting with a heterogeneous population of parasites when selecting for drug resistance in order to speed up the process. However, once the desired drug-resistant mutants have been obtained, their molecular genetic analysis is greatly facilitated if they all derive from the same parental, drug-sensitive clone. Otherwise, there are likely to be too many confounding nucleotide polymorphisms that are unrelated to the resistance phenotype. A typical procedure that facilitates downstream genomics, transcriptomics, and proteomics is to start the selection process with a fresh clone and select several lines independently. Thus, a melarsoprol-resistant mutant of T. b. rhodesiense was found to express only two genes (other than VSG) at a different level from the parental, melarsoprol-sensitive clone [16], even though it had taken two years of in vitro selection to obtain the mutant [73].
A successful approach towards high-level pentamidine resistance in T. brucei was to start with a genetically engineered clone that was already less susceptible to pentamidine since it was homozygously disrupted in the gene TbAT1, which encodes an aminopurine permease that also transports diamidine drugs [111]. This starting clone was already 2.4-fold resistant [112]; after several months of in vitro exposure to increasing concentrations of pentamidine, a resistance factor of 130 was obtained [72].

3.4. Selection with Mutagens vs. Adaptive Evolution

The use of chemical mutagens poses a similar dilemma as discussed above: it will increase the probability of obtaining a drug-resistant mutant and thus shorten the process of selection. At the same time, it will confront the downstream analyses of the obtained mutants with the challenging task of identifying which of the many mutations are the cause of drug resistance. Since back-crossing to the parental, drug-sensitive line is not feasible with trypanosomatid parasites, resistance selection based on chemical mutagenesis requires a large number of drug-resistant mutants that have been selected in parallel, preferably from the same, freshly cloned parent. Ethyl methanesulfonate (EMS) is frequently used as a mutagen to generate point mutations. It alkylates guanine to ethylguanine, which can form a base pair with thymine, leading to the transition from a G:C pair to A:T. EMS and other mutagens were applied to L. infantum promastigotes, followed by plating on media containing either miltefosine or paromomycin, which allowed the isolation of drug-resistant mutants [95]. Finally, the drug itself can be mutagenic. This is the case for ethidium bromide (homidium), which is used in veterinary medicine for T. congolense infections. Benznidazole, too, is mutagenic to trypanosomes [79].

3.5. Selection under Constant Pressure vs. Sudden Shock

The intuitive approach to select parasites for drug resistance in culture is to apply a steady selective pressure with a sublethal drug concentration, which can be gradually increased as the parasites lose susceptibility. This has been the most commonly used procedure (Table 3). Such an approach is likely to result in the accumulation of several mutations over time, and the phenotype of drug resistance that is ultimately obtained might result from a combination of genetic mechanisms. It is, therefore, imperative to freeze away intermediate samples, which will allow one to determine at what time point a given mutation has occurred. A different, potentially much faster approach is to start with a high inoculum of parasites, expose them to a supposedly lethal concentration of drug, and then wait and see whether, eventually, a population of parasites will recover. This worked well to select T. brucei for suramin resistance: when bloodstream-form trypanosomes were incubated with suramin at 5 to 25 fold the IC50, all cells seemed to be dead by the following day. However, the cultures were further incubated, and after 6 days a population had regrown that was about 100-fold resistant to suramin [51]. Obviously, such a shock treatment bears the risk that the culture is not going to recover simply because all the parasites are dead. Nevertheless, we think it is a worthwhile experiment to try, since it quickly delivers a (positive or negative) answer.

4. Biosafety Considerations and Conclusions

Parasites that have been selected for drug resistance may require additional biosafety measures or even an upgrade in the biosafety level. T. cruzi bears the highest biohazard risk among the trypanosomatids. Highly infectious not only by traumatic inoculation but also via mucous membranes, T. cruzi is considered in many countries as a pathogen of biosafety level 3 (Table 4). Only two drugs are available for the treatment of an accidental infection with T. cruzi, benznidazole and nifurtimox. Both are nitroimidazoles, and they have the same mechanism of action: activation by electron transfer catalyzed by nitroreductase 1 (NTR1), leading to the formation of radicals. Cross-resistance between benznidazole and nifurtimox due to reduced levels of NTR1 expression is the most frequently observed mechanism [61]. Thus, when T. cruzi is being selected for resistance to nitroimidazoles, this will demand even more stringent biosafety measures than required anyhow. This word of caution also applies to selection experiments with the insect stages, because densely grown cultures of epimastigote T. cruzi will contain infective metacyclic forms.
In summary, selecting trypanosomatid parasites for drug-resistant mutants requires special care. Moreover, whatever the experimental approach, it also requires patience and luck (in German Geduld and Glück), two of Paul Ehrlich’s famous Four Gs [113]. With points 3.1 to 3.5, we hope to have provided some guidance about the parameters that need to be considered when planning a drug selection experiment. While there is no bona fide best approach, a methodological consideration of the points outlined above will provide a framework for the successful planning of experiments.

Funding

This research was funded by the Swiss National Science Foundation, grant number CRSII5_183536.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are in the text.

Acknowledgments

We thank Monica Cal, Dennis Hauser, Sonja Keller-Märki and Romina Rocchetti for performing the assays for Table 2.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Murray, C.J.L.; Lopez, A.D. Measuring global health: Motivation and evolution of the Global Burden of Disease Study. Lancet 2017, 390, 1460–1464. [Google Scholar] [CrossRef]
  2. Lee, B.Y.; Bacon, K.M.; Bottazzi, M.E.; Hotez, P.J. Global economic burden of Chagas disease: A computational simulation model. Lancet Infect. Dis. 2013, 13, 342–348. [Google Scholar] [CrossRef] [Green Version]
  3. Barnes, R.L.; Shi, H.; Kolev, N.G.; Tschudi, C.; Ullu, E. Comparative genomics reveals two novel RNAi factors in Trypanosoma brucei and provides insight into the core machinery. PLoS Pathog. 2012, 8, e1002678. [Google Scholar] [CrossRef] [PubMed]
  4. Berriman, M.; Ghedin, E.; Hertz-Fowler, C.; Blandin, G.; Renauld, H.; Bartholomeu, D.C.; Lennard, N.J.; Caler, E.; Hamlin, N.E.; Haas, B.; et al. The genome of the African trypanosome Trypanosoma brucei. Science 2005, 309, 416–422. [Google Scholar] [CrossRef] [Green Version]
  5. El-Sayed, N.M.; Myler, P.J.; Bartholomeu, D.C.; Nilsson, D.; Aggarwal, G.; Tran, A.N.; Ghedin, E.; Worthey, E.A.; Delcher, A.L.; Blandin, G.; et al. The genome sequence of Trypanosoma cruzi, etiologic agent of Chagas disease. Science 2005, 309, 409–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Downing, T.; Imamura, H.; Decuypere, S.; Clark, T.G.; Coombs, G.H.; Cotton, J.A.; Hilley, J.D.; de Doncker, S.; Maes, I.; Mottram, J.C.; et al. Whole genome sequencing of multiple Leishmania donovani clinical isolates provides insights into population structure and mechanisms of drug resistance. Genome Res. 2011, 21, 2143–2156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Patrick, K.L.; Shi, H.; Kolev, N.G.; Ersfeld, K.; Tschudi, C.; Ullu, E. Distinct and overlapping roles for two Dicer-like proteins in the RNA interference pathways of the ancient eukaryote Trypanosoma brucei. Proc. Natl. Acad. Sci. USA 2009, 106, 17933–17938. [Google Scholar] [CrossRef] [Green Version]
  8. Silva, M.R.G.; Tosar, J.P.; Frugier, M.; Pantano, S.; Bonilla, B.; Esteban, L.; Serra, E.; Rovira, C.; Robello, C.; Cayota, A. Cloning, characterization and subcellular localization of a Trypanosoma cruzi argonaute protein defining a new subfamily distinctive of trypanosomatids. Gene 2010, 466, 26–35. [Google Scholar] [CrossRef]
  9. Lye, L.-F.; Owens, K.; Shi, H.; Murta, S.M.F.; Vieira, A.C.; Turco, S.J.; Tschudi, C.; Ullu, E.; Beverley, S.M. Retention and loss of RNA interference pathways in trypanosomatid protozoans. PLoS Pathog. 2010, 6, e1001161. [Google Scholar] [CrossRef] [Green Version]
  10. Batista, T.M.; Marques, J.T. RNAi pathways in parasitic protists and worms. J. Proteom. 2011, 74, 1504–1514. [Google Scholar] [CrossRef]
  11. Beneke, T.; Madden, R.; Makin, L.; Valli, J.; Sunter, J.; Gluenz, E. A CRISPR Cas9 high-throughput genome editing toolkit for kinetoplastids. R. Soc. Open Sci. 2017, 4, 170095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Peng, D.; Kurup, S.P.; Yao, P.Y.; Minning, T.A.; Tarleton, R.L. CRISPR-Cas9-mediated single-gene and gene family disruption in Trypanosoma cruzi. mBio 2014, 6, e02097-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lander, N.; Li, Z.H.; Niyogi, S.; Docampo, R. CRISPR/Cas9-Induced Disruption of Paraflagellar Rod Protein 1 and 2 Genes in Trypanosoma cruzi Reveals Their Role in Flagellar Attachment. mBio 2015, 6, e01012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Zhang, W.W.; Matlashewski, G. CRISPR-Cas9-Mediated Genome Editing in Leishmania donovani. mBio 2015, 6, e00861. [Google Scholar] [CrossRef] [Green Version]
  15. Sangster, N.C.; Song, J.; Demeler, J. Resistance as a tool for discovering and understanding targets in parasite neuromusculature. Parasitology 2005, 131, S179–S190. [Google Scholar] [CrossRef]
  16. Graf, F.E.; Ludin, P.; Arquint, C.; Schmidt, R.S.; Schaub, N.; Renggli, C.K.; Munday, J.C.; Krezdorn, J.; Baker, N.; Horn, D.; et al. Comparative genomics of drug resistance in Trypanosoma brucei rhodesiense. Cell. Mol. Life Sci. 2016, 73, 3387–3400. [Google Scholar] [CrossRef] [Green Version]
  17. Leprohon, P.; Fernandez-Prada, C.; Gazanion, E.; Monte-Neto, R.; Ouellette, M. Drug resistance analysis by next generation sequencing in Leishmania. Int. J. Parasitol. Drugs Drug Resist. 2015, 5, 26–35. [Google Scholar] [CrossRef] [Green Version]
  18. Alsford, S.; Eckert, S.; Baker, N.; Glover, L.; Sanchez-Flores, A.; Leung, K.F.; Turner, D.J.; Field, M.C.; Berriman, M.; Horn, D. High-throughput decoding of antitrypanosomal drug efficacy and resistance. Nature 2012, 482, 232–236. [Google Scholar] [CrossRef] [Green Version]
  19. Alsford, S.; Turner, D.J.; Obado, S.O.; Sanchez-Flores, A.; Glover, L.; Berriman, M.; Hertz-Fowler, C.; Horn, D. High-throughput phenotyping using parallel sequencing of RNA interference targets in the African trypanosome. Genome Res. 2011, 21, 915–924. [Google Scholar] [CrossRef] [Green Version]
  20. Schumann Burkard, G.; Jutzi, P.; Roditi, I. Genome-wide RNAi screens in bloodstream form trypanosomes identify drug transporters. Mol. Biochem. Parasitol. 2011, 175, 91–94. [Google Scholar] [CrossRef]
  21. Baker, N.; Alsford, S.; Horn, D. Genome-wide RNAi screens in African trypanosomes identify the nifurtimox activator NTR and the eflornithine transporter AAT6. Mol. Biochem. Parasitol. 2011, 176, 55–57. [Google Scholar] [CrossRef] [PubMed]
  22. Kuettel, S.; Greenwald, J.; Kostrewa, D.; Ahmed, S.; Scapozza, L.; Perozzo, R. Crystal structures of T. b. rhodesiense adenosine kinase complexed with inhibitor and activator: Implications for catalysis and hyperactivation. PLoS Negl. Trop. Dis. 2011, 5, e1164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kuettel, S.; Mosimann, M.; Maser, P.; Kaiser, M.; Brun, R.; Scapozza, L.; Perozzo, R. Adenosine Kinase of T. b. Rhodesiense identified as the putative target of 4-[5-(4-phenoxyphenyl)-2H-pyrazol-3-yl]morpholine using chemical proteomics. PLoS Negl. Trop. Dis. 2009, 3, e506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Matveyev, A.V.; Alves, J.M.; Serrano, M.G.; Lee, V.; Lara, A.M.; Barton, W.A.; Costa-Martins, A.G.; Beverley, S.M.; Camargo, E.P.; Teixeira, M.M.; et al. The Evolutionary Loss of RNAi Key Determinants in Kinetoplastids as a Multiple Sporadic Phenomenon. J. Mol. Evol. 2017, 84, 104–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Soares Medeiros, L.C.; South, L.; Peng, D.; Bustamante, J.M.; Wang, W.; Bunkofske, M.; Perumal, N.; Sanchez-Valdez, F.; Tarleton, R.L. Rapid, Selection-Free, High-Efficiency Genome Editing in Protozoan Parasites Using CRISPR-Cas9 Ribonucleoproteins. mBio 2017, 8, 6. [Google Scholar] [CrossRef] [Green Version]
  26. Sollelis, L.; Ghorbal, M.; MacPherson, C.R.; Martins, R.M.; Kuk, N.; Crobu, L.; Bastien, P.; Scherf, A.; Lopez-Rubio, J.J.; Sterkers, Y. First efficient CRISPR-Cas9-mediated genome editing in Leishmania parasites. Cell. Microbiol. 2015, 17, 1405–1412. [Google Scholar] [CrossRef] [Green Version]
  27. Rico, E.; Jeacock, L.; Kovarova, J.; Horn, D. Inducible high-efficiency CRISPR-Cas9-targeted gene editing and precision base editing in African trypanosomes. Sci. Rep. 2018, 8, 7960. [Google Scholar] [CrossRef] [Green Version]
  28. Shaw, S.; Knusel, S.; Hoenner, S.; Roditi, I. A transient CRISPR/Cas9 expression system for genome editing in Trypanosoma brucei. BMC Res. Notes 2020, 13, 268. [Google Scholar] [CrossRef]
  29. Beneke, T.; Gluenz, E. Bar-seq strategies for the LeishGEdit toolbox. Mol. Biochem. Parasitol. 2020, 239, 111295. [Google Scholar] [CrossRef]
  30. Yagoubat, A.; Corrales, R.M.; Bastien, P.; Lévêque, M.F.; Sterkers, Y. Gene Editing in Trypanosomatids: Tips and Tricks in the CRISPR-Cas9 Era. Trends Parasitol. 2020, 36, 745–760. [Google Scholar] [CrossRef]
  31. Fernandez-Prada, C.; Sharma, M.; Plourde, M.; Bresson, E.; Roy, G.; Leprohon, P.; Ouellette, M. High-throughput Cos-Seq screen with intracellular Leishmania infantum for the discovery of novel drug-resistance mechanisms. Int. J. Parasitol. Drugs Drug Resist. 2018, 8, 165–173. [Google Scholar] [CrossRef] [PubMed]
  32. Gazanion, É.; Fernández-Prada, C.; Papadopoulou, B.; Leprohon, P.; Ouellette, M. Cos-Seq for high-throughput identification of drug target and resistance mechanisms in the protozoan parasite Leishmania. Proc. Natl. Acad. Sci. USA 2016, 113, E3012–E3021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rajao, M.A.; Furtado, C.; Alves, C.L.; Passos-Silva, D.G.; de Moura, M.B.; Schamber-Reis, B.L.; Kunrath-Lima, M.; Zuma, A.A.; Vieira-da-Rocha, J.P.; Garcia, J.B.; et al. Unveiling benznidazole’s mechanism of action through overexpression of DNA repair proteins in Trypanosoma cruzi. Environ. Mol. Mutagen. 2014, 55, 309–321. [Google Scholar] [CrossRef] [PubMed]
  34. Wall, R.J.; Rico, E.; Lukac, I.; Zuccotto, F.; Elg, S.; Gilbert, I.H.; Freund, Y.; Alley, M.R.K.; Field, M.C.; Wyllie, S.; et al. Clinical and veterinary trypanocidal benzoxaboroles target CPSF3. Proc. Natl. Acad. Sci. USA 2018, 115, 9616–9621. [Google Scholar] [CrossRef] [Green Version]
  35. Sioud, M. (Ed.) Main Approaches to Target Discovery and Validation. In Target Discovery and Validation Reviews and Protocols. Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2007; Volume 360, pp. 1–12. [Google Scholar]
  36. Werbovetz, K.A. Target-Based Drug Discovery for Malaria, Leishmaniasis, and Trypanosomiasis. Curr. Med. Chem. 2000, 7, 835–860. [Google Scholar] [CrossRef]
  37. Kubota, K.; Funabashi, M.; Ogura, Y. Target deconvolution from phenotype-based drug discovery by using chemical proteomics approaches. Biochim. Biophys. Acta Proteins Proteom. 2019, 1867, 22–27. [Google Scholar] [CrossRef]
  38. Mercer, L.; Bowling, T.; Perales, J.; Freeman, J.; Nguyen, T.; Bacchi, C.; Yarlett, N.; Don, R.; Jacobs, R.; Nare, B. 2,4-Diaminopyrimidines as potent inhibitors of Trypanosoma brucei and identification of molecular targets by a chemical proteomics approach. PLoS Negl. Trop. Dis. 2011, 5, e956. [Google Scholar] [CrossRef] [Green Version]
  39. Yang, P.Y.; Wang, M.; He, C.Y.; Yao, S.Q. Proteomic profiling and potential cellular target identification of K11777, a clinical cysteine protease inhibitor, in Trypanosoma brucei. Chem. Commun. 2012, 48, 835–837. [Google Scholar] [CrossRef]
  40. Yang, P.Y.; Wang, M.; Liu, K.; Ngai, M.H.; Sheriff, O.; Lear, M.J.; Sze, S.K.; He, C.Y.; Yao, S.Q. Parasite-based screening and proteome profiling reveal orlistat, an FDA-approved drug, as a potential anti Trypanosoma brucei agent. Chemistry 2012, 18, 8403–8413. [Google Scholar] [CrossRef]
  41. Chidley, C.; Haruki, H.; Pedersen, M.G.; Muller, E.; Johnsson, K. A yeast-based screen reveals that sulfasalazine inhibits tetrahydrobiopterin biosynthesis. Nat. Chem. Biol. 2011, 7, 375–383. [Google Scholar] [CrossRef]
  42. Terstappen, G.C.; Schlupen, C.; Raggiaschi, R.; Gaviraghi, G. Target deconvolution strategies in drug discovery. Nat. Rev. Drug Discov 2007, 6, 891–903. [Google Scholar] [CrossRef] [PubMed]
  43. Ehrlich, P. Chemotherapeutische Trypanosomen-Studien. Berl. klin. Wochenschr. 1907, 11, 310–314. [Google Scholar]
  44. Fleming, A. Penicillin. Nobel Lecture 1945. Nobel Foundation. Available online: www.nobelprize.org (accessed on 22 December 2021).
  45. Ehrlich, P. Partial cell functions. Nobel Lecture 1908. Nobel Foundation. Available online: www.nobelprize.org (accessed on 22 December 2021).
  46. Chugh, M.; Scheurer, C.; Sax, S.; Bilsland, E.; van Schalkwyk, D.A.; Wicht, K.J.; Hofmann, N.; Sharma, A.; Bashyam, S.; Singh, S.; et al. Identification and deconvolution of cross-resistance signals from antimalarial compounds using multidrug-resistant Plasmodium falciparum strains. Antimicrob. Agents Chemother. 2015, 59, 1110–1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Kaminsky, R.; Mäser, P. Drug resistance in African trypanosomes. Curr. Op. Anti Inf. Inv. Drugs 2000, 2, 76–82. [Google Scholar]
  48. Scott, A.G.; Tait, A.; Turner, M.R. Characterisation of cloned lines of Trypanosoma brucei expressing stable resistance to MelCy and Suramin. Acta Trop. 1996, 60, 251–262. [Google Scholar] [CrossRef]
  49. Scott, A.G.; Tait, A.; Turner, C.M.R. Trypanosoma brucei: Lack of cross-resistance to melarsoprol in vitro by cymelarsan-resistant parasites. Exp. Parasitol. 1997, 86, 181–190. [Google Scholar] [CrossRef]
  50. Keiser, J.; Ericsson, O.; Burri, C. Investigations of the metabolites of the trypanocidal drug melarsoprol. Clin. Pharmacol. Ther. 2000, 67, 478–488. [Google Scholar] [CrossRef]
  51. Wiedemar, N.; Graf, F.E.; Zwyer, M.; Ndomba, E.; Kunz Renggli, C.; Cal, M.; Schmidt, R.S.; Wenzler, T.; Mäser, P. Beyond immune escape: A variant surface glycoprotein causes suramin resistance in Trypanosoma brucei. Mol. Microbiol. 2018, 107, 57–67. [Google Scholar] [CrossRef] [Green Version]
  52. Yorke, W.; Murgatroyd, F. The action in vitro of certain arsenical and antimonial compounds on T. rhodesiense and on atoxyl- and acriflavine-resistant strains of this parasite. Ann. Trop. Med. Parasitol. 1930, 24, 449–476. [Google Scholar] [CrossRef]
  53. Yorke, W.; Murgatroyd, F.; Hawking, F. Preliminary contribution on the nature of drug resistance. Ann. Trop. Med. Parasitol. 1931, 25, 351–358. [Google Scholar] [CrossRef]
  54. Yorke, W.; Murgatroyd, F.; Hawking, F. Comparison of Strains of T. rhodesiense Made Resistant to Various Arsenicals and Antimonials, to Bayer 205, and to Acriflavine, Respectively. Ann. Trop. Med. Parasitol. 1933, 26, 577–586. [Google Scholar] [CrossRef]
  55. Lourie, E.M.; Yorke, W. The Preparation of Strains of Trypanosomes Resistant to Synthalin and Undecane Diamidine, and an Analysis of Their Characters. Ann. Trop. Med. Parasitol. 1938, 32, 201–213. [Google Scholar] [CrossRef]
  56. Fulton, J.D.; Yorke, W. Further Observations on the Stability of Drug-Resistance in Trypanosomes. Ann. Trop. Med. Parasitol. 1942, 35, 221–227. [Google Scholar] [CrossRef]
  57. Rollo, I.M.; Williamson, J. Acquired resistance to ‘melarsen’, tryparsamide and amidines in pathogenic trypanosomes after treatment with ‘melarsen’ alone. Nature 1951, 167, 147–148. [Google Scholar] [CrossRef]
  58. Frommel, T.O.; Balber, A.E. Flow cytofluorimetric analysis of drug accumulation by multidrug-resistant Trypanosoma brucei brucei and T. b. rhodesiense. Mol. Biochem. Parasitol. 1987, 26, 183–192. [Google Scholar] [CrossRef]
  59. Pospichal, H.; Brun, R.; Kaminsky, R.; Jenni, L. Induction of resistance to melarsenoxide cysteamine (Mel Cy) in Trypanosoma brucei brucei. Acta Trop. 1994, 58, 187–197. [Google Scholar] [CrossRef]
  60. Fairlamb, A.H.; Carter, N.S.; Cunningham, M.; Smith, K. Characterisation of melarsen-resistant Trypanosoma brucei brucei with respect to cross-resistance to other drugs and trypanothione metabolism. Mol. Biochem. Parasitol. 1992, 53, 213–222. [Google Scholar] [CrossRef]
  61. Murta, S.M.; Romanha, A.J. In vivo selection of a population of Trypanosoma cruzi and clones resistant to benznidazole. Parasitology 1998, 116, 165–171. [Google Scholar] [CrossRef]
  62. Dos Santos, F.M.; Caldas, S.; de Assis Cau, S.B.; Crepalde, G.P.; de Lana, M.; Machado-Coelho, G.L.; Veloso, V.M.; Bahia, M.T. Trypanosoma cruzi: Induction of benznidazole resistance in vivo and its modulation by in vitro culturing and mice infection. Exp. Parasitol. 2008, 120, 385–390. [Google Scholar] [CrossRef]
  63. Hendrickx, S.; Mondelaers, A.; Eberhardt, E.; Delputte, P.; Cos, P.; Maes, L. In Vivo Selection of Paromomycin and Miltefosine Resistance in Leishmania donovani and L. infantum in a Syrian Hamster Model. Antimicrob. Agents Chemother. 2015, 59, 4714–4718. [Google Scholar] [CrossRef] [Green Version]
  64. Perry, M.R.; Wyllie, S.; Raab, A.; Feldmann, J.; Fairlamb, A.H. Chronic exposure to arsenic in drinking water can lead to resistance to antimonial drugs in a mouse model of visceral leishmaniasis. Proc. Natl. Acad. Sci. USA 2013, 110, 19932–19937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lanteri, C.A.; Stewart, M.L.; Brock, J.M.; Alibu, V.P.; Meshnick, S.R.; Tidwell, R.R.; Barrett, M.P. Roles for the Trypanosoma brucei P2 transporter in DB75 uptake and resistance. Mol. Pharmacol. 2006, 70, 1585–1592. [Google Scholar] [CrossRef] [PubMed]
  66. Teka, I.A.; Kazibwe, A.J.; El-Sabbagh, N.; Al-Salabi, M.I.; Ward, C.P.; Eze, A.A.; Munday, J.C.; Mäser, P.; Matovu, E.; Barrett, M.P.; et al. The diamidine diminazene aceturate is a substrate for the high-affinity pentamidine transporter: Implications for the development of high resistance levels in trypanosomes. Mol. Pharmacol. 2011, 80, 110–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Vincent, I.M.; Creek, D.; Watson, D.G.; Kamleh, M.A.; Woods, D.J.; Wong, P.E.; Burchmore, R.J.; Barrett, M.P. A molecular mechanism for eflornithine resistance in African trypanosomes. PLoS Pathog. 2010, 6, e1001204. [Google Scholar] [CrossRef] [Green Version]
  68. Ranade, R.M.; Gillespie, J.R.; Shibata, S.; Verlinde, C.L.; Fan, E.; Hol, W.G.; Buckner, F.S. Induced resistance to methionyl-tRNA synthetase inhibitors in Trypanosoma brucei is due to overexpression of the target. Antimicrob. Agents Chemother. 2013, 57, 3021–3028. [Google Scholar] [CrossRef] [Green Version]
  69. Wilson, K.; Berens, R.L.; Sifri, C.D.; Ullman, B. Amplification of the inosinate dehydrogenase gene in Trypanosoma brucei gambiense due to an increase in chromosome copy number. J. Biol. Chem. 1994, 269, 28979–28987. [Google Scholar] [CrossRef]
  70. Sokolova, A.Y.; Wyllie, S.; Patterson, S.; Oza, S.L.; Read, K.D.; Fairlamb, A.H. Cross-resistance to nitro drugs and implications for treatment of human African trypanosomiasis. Antimicrob. Agents Chemother. 2010, 54, 2893–2900. [Google Scholar] [CrossRef] [Green Version]
  71. Berger, B.J.; Carter, N.S.; Fairlamb, A.H. Polyamine and pentamidine metabolism in African trypanosomes. Acta Trop. 1993, 54, 215–224. [Google Scholar] [CrossRef]
  72. Bridges, D.J.; Gould, M.K.; Nerima, B.; Mäser, P.; Burchmore, R.J.; de Koning, H.P. Loss of the high-affinity pentamidine transporter is responsible for high levels of cross-resistance between arsenical and diamidine drugs in African trypanosomes. Mol. Pharmacol. 2007, 71, 1098–1108. [Google Scholar] [CrossRef] [Green Version]
  73. Bernhard, S.C.; Nerima, B.; Mäser, P.; Brun, R. Melarsoprol- and pentamidine-resistant Trypanosoma brucei rhodesiense populations and their cross-resistance. Int. J. Parasitol. 2007, 37, 1443–1448. [Google Scholar] [CrossRef]
  74. Ali, J.A.; Creek, D.J.; Burgess, K.; Allison, H.C.; Field, M.C.; Maser, P.; De Koning, H.P. Pyrimidine salvage in Trypanosoma brucei bloodstream forms and the trypanocidal action of halogenated pyrimidines. Mol. Pharmacol. 2013, 83, 439–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Nirde, P.; Larroque, C.; Barnabe, C. Drug-resistant epimastigotes of Trypanosoma cruzi and persistence of this phenotype after differentiation into amastigotes. C. R. Acad. Sci. III 1995, 318, 1239–1244. [Google Scholar] [PubMed]
  76. Campos, M.C.; Castro-Pinto, D.B.; Ribeiro, G.A.; Berredo-Pinho, M.M.; Gomes, L.H.; da Silva Bellieny, M.S.; Goulart, C.M.; Echevarria, A.; Leon, L.L. P-glycoprotein efflux pump plays an important role in Trypanosoma cruzi drug resistance. Parasitol. Res. 2013, 112, 2341–2351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Campos, M.C.; Leon, L.L.; Taylor, M.C.; Kelly, J.M. Benznidazole-resistance in Trypanosoma cruzi: Evidence that distinct mechanisms can act in concert. Mol. Biochem. Parasitol. 2014, 193, 17–19. [Google Scholar] [CrossRef]
  78. Mejia, A.M.; Hall, B.S.; Taylor, M.C.; Gomez-Palacio, A.; Wilkinson, S.R.; Triana-Chavez, O.; Kelly, J.M. Benznidazole-resistance in Trypanosoma cruzi is a readily acquired trait that can arise independently in a single population. J. Infect. Dis. 2012, 206, 220–228. [Google Scholar] [CrossRef] [Green Version]
  79. Campos, M.C.; Phelan, J.; Francisco, A.F.; Taylor, M.C.; Lewis, M.D.; Pain, A.; Clark, T.G.; Kelly, J.M. Genome-wide mutagenesis and multi-drug resistance in American trypanosomes induced by the front-line drug benznidazole. Sci. Rep. 2017, 7, 14407. [Google Scholar] [CrossRef] [Green Version]
  80. Nogueira, F.B.; Krieger, M.A.; Nirde, P.; Goldenberg, S.; Romanha, A.J.; Murta, S.M. Increased expression of iron-containing superoxide dismutase-A (TcFeSOD-A) enzyme in Trypanosoma cruzi population with in vitro-induced resistance to benznidazole. Acta Trop. 2006, 100, 119–132. [Google Scholar] [CrossRef]
  81. Buckner, F.S.; Wilson, A.J.; White, T.C.; Van Voorhis, W.C. Induction of resistance to azole drugs in Trypanosoma cruzi. Antimicrob. Agents Chemother. 1998, 42, 3245–3250. [Google Scholar] [CrossRef] [Green Version]
  82. Wilkinson, S.R.; Taylor, M.C.; Horn, D.; Kelly, J.M.; Cheeseman, I. A mechanism for cross-resistance to nifurtimox and benznidazole in trypanosomes. Proc. Natl. Acad. Sci. USA 2008, 105, 5022–5027. [Google Scholar] [CrossRef] [Green Version]
  83. Nozaki, T.; Engel, J.C.; Dvorak, J.A. Cellular and molecular biological analyses of nifurtimox resistance in Trypanosoma cruzi. Am. J. Trop. Med. Hyg. 1996, 55, 111–117. [Google Scholar] [CrossRef]
  84. Nozaki, T.; Dvorak, J.A. Molecular biology studies of tubercidin resistance in Trypanosoma cruzi. Parasitol. Res. 1993, 79, 451–455. [Google Scholar] [CrossRef] [PubMed]
  85. Garvey, E.P.; Coderre, J.A.; Santi, D.V. Selection and properties of Leishmania tropica resistant to 10-propargyl-5,8-dideazafolate, an inhibitor of thymidylate synthetase. Mol. Biochem. Parasitol. 1985, 17, 79–91. [Google Scholar] [CrossRef]
  86. Yasur-Landau, D.; Jaffe, C.L.; Doron-Faigenboim, A.; David, L.; Baneth, G. Induction of allopurinol resistance in Leishmania infantum isolated from dogs. PLoS Negl. Trop. Dis. 2017, 11, e0005910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Garcia-Hernandez, R.; Manzano, J.I.; Castanys, S.; Gamarro, F. Leishmania donovani develops resistance to drug combinations. PLoS Negl. Trop. Dis. 2012, 6, e1974. [Google Scholar] [CrossRef] [Green Version]
  88. Beverley, S.M.; Coderre, J.A.; Santi, D.V.; Schimke, R.T. Unstable DNA amplifications in methotrexate-resistant Leishmania consist of extrachromosomal circles which relocalize during stabilization. Cell 1984, 38, 431–439. [Google Scholar] [CrossRef]
  89. Detke, S.; Katakura, K.; Chang, K.P. DNA amplification in arsenite-resistant Leishmania. Exp. Cell. Res. 1989, 180, 161–170. [Google Scholar] [CrossRef]
  90. Marquis, N.; Gourbal, B.; Rosen, B.P.; Mukhopadhyay, R.; Ouellette, M. Modulation in aquaglyceroporin AQP1 gene transcript levels in drug-resistant Leishmania. Mol. Microbiol. 2005, 57, 1690–1699. [Google Scholar] [CrossRef]
  91. Chiquero, M.J.; Perez-Victoria, J.M.; O’Valle, F.; Gonzalez-Ros, J.M.; del Moral, R.G.; Ferragut, J.A.; Castanys, S.; Gamarro, F. Altered drug membrane permeability in a multidrug-resistant Leishmania tropica line. Biochem. Pharmacol. 1998, 55, 131–139. [Google Scholar] [CrossRef]
  92. Kaur, K.; Coons, T.; Emmett, K.; Ullman, B. Methotrexate-resistant Leishmania donovani genetically deficient in the folate-methotrexate transporter. J. Biol. Chem. 1988, 263, 7020–7028. [Google Scholar] [CrossRef]
  93. Iovannisci, D.M.; Beverley, S.M. Structural alterations of chromosome 2 in Leishmania major as evidence for diploidy, including spontaneous amplification of the mini-exon array. Mol. Biochem. Parasitol. 1989, 34, 177–188. [Google Scholar] [CrossRef]
  94. Seifert, K.; Matu, S.; Javier Perez-Victoria, F.; Castanys, S.; Gamarro, F.; Croft, S.L. Characterisation of Leishmania donovani promastigotes resistant to hexadecylphosphocholine (miltefosine). Int. J. Antimicrob. Agents 2003, 22, 380–387. [Google Scholar] [CrossRef]
  95. Bhattacharya, A.; Leprohon, P.; Bigot, S.; Padmanabhan, P.K.; Mukherjee, A.; Roy, G.; Gingras, H.; Mestdagh, A.; Papadopoulou, B.; Ouellette, M. Coupling chemical mutagenesis to next generation sequencing for the identification of drug resistance mutations in Leishmania. Nat. Commun. 2019, 10, 5627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Jhingran, A.; Chawla, B.; Saxena, S.; Barrett, M.P.; Madhubala, R. Paromomycin: Uptake and resistance in Leishmania donovani. Mol. Biochem. Parasitol. 2009, 164, 111–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Ullman, B.; Carrero-Valenzuela, E.; Coons, T. Leishmania donovani: Isolation and characterization of sodium stibogluconate (Pentostam)-resistant cell lines. Exp. Parasitol. 1989, 69, 157–163. [Google Scholar] [CrossRef]
  98. Ellenberger, T.E.; Beverley, S.M. Multiple drug resistance and conservative amplification of the H region in Leishmania major. J. Biol. Chem. 1989, 264, 15094–15103. [Google Scholar] [CrossRef]
  99. Alzahrani, K.J.H.; Ali, J.A.M.; Eze, A.A.; Looi, W.L.; Tagoe, D.N.A.; Creek, D.J.; Barrett, M.P.; de Koning, H.P. Functional and genetic evidence that nucleoside transport is highly conserved in Leishmania species: Implications for pyrimidine-based chemotherapy. Int. J. Parasitol. Drugs Drug Resist. 2017, 7, 206–226. [Google Scholar] [CrossRef] [PubMed]
  100. Hawking, F.; Walker, P.J. Analysis of the development of arsenical resistance in trypanosomes in vitro. Exp. Parasitol. 1966, 18, 63–86. [Google Scholar] [CrossRef]
  101. Brun, R.; Schönenberger, M. Cultivation and in vivo cloning of procyclic culture forms of Trypanosoma brucei in a semi-defined medium. Acta Trop. 1979, 36, 289–292. [Google Scholar]
  102. Berens, R.L.; Brun, R.; Krassner, S.M. A simple monophasic medium for axenic culture of hemoflagellates. J. Parasitol. 1976, 62, 360–365. [Google Scholar] [CrossRef]
  103. Krassner, S.M.; Flory, B. Proline metabolism in Leishmania donovani promastigotes. J. Protozool. 1972, 19, 682–685. [Google Scholar] [CrossRef]
  104. Baltz, T.; Baltz, D.; Giroud, C.; Crockett, J. Cultivation in a semi-defined medium of animal infective forms of Trypanosoma brucei, T. equiperdum, T. evansi, T. rhodesiense and T. gambiense. EMBO J. 1985, 4, 1273–1277. [Google Scholar] [CrossRef] [PubMed]
  105. Balanco, J.M.; Pral, E.M.; da Silva, S.; Bijovsky, A.T.; Mortara, R.A.; Alfieri, S.C. Axenic cultivation and partial characterization of Leishmania braziliensis amastigote-like stages. Parasitology 1998, 116, 103–113. [Google Scholar] [CrossRef] [PubMed]
  106. Engel, J.C.; Dvorak, J.A. Trypanosoma cruzi: Cell biological behavior of epimastigote and amastigote forms in axenic culture. J. Protozool. 1988, 35, 513–518. [Google Scholar] [CrossRef]
  107. Revollo, S.; Oury, B.; Vela, A.; Tibayrenc, M.; Sereno, D. In Vitro Benznidazole and Nifurtimox Susceptibility Profile of Trypanosoma cruzi Strains Belonging to Discrete Typing Units TcI, TcII, and TcV. Pathogens 2019, 8, 197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. de Koning, H. Uptake of pentamidine in Trypanosoma brucei is mediated by three distinct transporters: Implications for cross-resistance with arsenicals. Mol. Pharmacol. 2001, 59, 586–592. [Google Scholar] [CrossRef] [Green Version]
  109. Ziegelbauer, K.; Overath, P. Identification of invariant surface glycoproteins in the bloodstream stage of Trypanosoma brucei. J. Biol. Chem. 1992, 267, 10791–10796. [Google Scholar] [CrossRef]
  110. Nyeko, J.H.; Golder, T.K.; Otieno, L.H. Selection for drug resistance in Trypanosoma congolense during cyclic transmissions through Glossina morsitans morsitans and drug treated rabbits. Acta Trop. 1988, 45, 21–26. [Google Scholar]
  111. Mäser, P.; Sütterlin, C.; Kralli, A.; Kaminsky, R. A nucleoside transporter from Trypanosoma brucei involved in drug resistance. Science 1999, 285, 242–244. [Google Scholar] [CrossRef]
  112. Matovu, E.; Stewart, M.L.; Geiser, F.; Brun, R.; Mäser, P.; Wallace, L.J.; Burchmore, R.J.; Enyaru, J.C.; Barrett, M.P.; Kaminsky, R.; et al. Mechanisms of arsenical and diamidine uptake and resistance in Trypanosoma brucei. Eukaryot. Cell 2003, 2, 1003–1008. [Google Scholar] [CrossRef] [Green Version]
  113. Raju, T.N.M. The Nobel Chronicles. 1908: Elie Metchnikoff (1845–1916) and Paul Ehrlich (1854–1915). Lancet 1998, 352, 661. [Google Scholar] [CrossRef]
Table 1. Molecular and cellular characteristics of the three selected trypanosomatids.
Table 1. Molecular and cellular characteristics of the three selected trypanosomatids.
T. bruceiT. cruziL. donovani
Genome size [4,5,6]26.1 Mb60.4 Mb32.4 Mb
Protein-coding genes [4,5,6]9068~12,000>8000
Genes of RNAi pathway [7,8,9,10]presentpartially presentabsent
RNAi gene silencing [7,8,9,10]functionalnon-functionalnon-functional
CRISPR/Cas9 editing [11,12,13,14]establishedestablishedestablished
Mammalian stagesextracell. trypomastigotesintracell. amastigote,
extracell. trypomastigote
intracell. amastigote
Vector stagesprocyclic trypomastigote,
epimastigote,
metacyclic trypomastigote
procyclic epimastigote,
metacyclic trypomastigote
procyclic promastigote,
metacyclic promastigote
Table 2. Standard drugs and sensitivity of mammalian vs. insect stages. All values are in vitro IC50 in µg/ml, original data from our trypanosomatid drug screening platform.
Table 2. Standard drugs and sensitivity of mammalian vs. insect stages. All values are in vitro IC50 in µg/ml, original data from our trypanosomatid drug screening platform.
ParasiteDrugMammalian Stage
Intracellular
Mammalian Stage
Axenic
Vector Stage
T. bruceiPentamidinen.a.0.0010.43
Suraminn.a.0.056>10
Melarsoproln.a.0.0040.057
Eflornithinen.a.2.0>100
Nifurtimoxn.a.0.311.6
Fexinidazolen.a.0.621.2
T. cruziBenznidazole0.47n.a.3.1
Nifurtimox0.14n.a.0.87
L. donovaniPentostam92220>1000
Miltefosine1.40.293.8
Amphotericin B0.330.260.03
Paromomycin28>3010
Table 3. Reports of successful in vitro selection of trypanosomatids for drug resistance (BSF, bloodstream form; PCF, procyclic form; epi, epimastigotes; pro, promastigotes; trypo, trypomastigotes; RF, resistance factor; n.s., not specified).
Table 3. Reports of successful in vitro selection of trypanosomatids for drug resistance (BSF, bloodstream form; PCF, procyclic form; epi, epimastigotes; pro, promastigotes; trypo, trypomastigotes; RF, resistance factor; n.s., not specified).
DrugSpeciesStageMutagenPressureDurationRFRef.
DB75T. b. bruceiBSFnosteady2.5 mth20[65]
BerenilT. b. brucei Δat1BSFnosteady5 mth9.2[66]
EflornithineT. b. bruceiBSFnosteady2 mth41[67]
Eflornithine, pentamidine, 1433T. b. bruceiBSFnosteady50–120 d32[68]
Melarsenoxide cysteamineT. b. bruceiBSFnosteady4 mth15[59]
Mycophenolic acidT. b. gambiensePCFnosteadyn.s.17[69]
NifurtimoxT. b. bruceiBSFnosteady4.7 mth8[70]
PentamidineT. b. bruceiBSFnosteady2 mth26[71]
PentamidineT. b. brucei Δat1BSFnosteadyseveral mth130[72]
Pentamidine, melarsoprolT. b. rhodesienseBSFnosteady21 mth140, 24[73]
Pyrimidine analogsT. b. bruceiBSFnosteadyseveral mth83–830[74]
SuraminT. b. rhodesienseBSFnoshock6 d96[51]
BenznidazoleT. cruziepinosteadyn.s.26[75]
BenznidazoleT. cruziepinointermittent15 w≥4.7[76]
BenznidazoleT. cruziepinosteadyn.s.9-26[77]
BenznidazoleT. cruziepinosteadyseveral wn.s.[78]
BenznidazoleT. cruziepinosteady4 mth9–26[79]
BenznidazoleT. cruziepinosteadyn.s.23[80]
FluconazoleT. cruziepinosteady4 mth100[81]
NifurtimoxT. cruziepinosteady8 mth4[82]
NifurtimoxT. cruziepi, tryponosteady60 d3–10[83]
TubercidinT. cruziepiyesshock1 mth180–260[84]
CB3717L. tropicapronosteady3–12 mth25000[85]
AllopurinolL. infantumpronosteady23 w20[86]
Amphotericin B, miltefosine
paromomycin, SbIII
L. donovanipronosteady18 w11–20[87]
MethotrexateL. tropicapronosteady3–11 mthn.s.[88]
ArseniteL. mex., L. amazon.pronosteady1 mth12[89]
Hoechst 33342L. donovanipronosteadyn.s.30[90]
DaunomycinL. tropicapronosteady6 mth62[91]
MethotrexateL. donovanipronoshock7–10 genn.s.[92]
MethotrexateL. majorpronosteadyn.s.n.s.[93]
MiltefosineL. donovanipronosteady6 mth15[94]
Miltefosine, paromomycinL. infantumproyessteady10 d2.5–8.5[95]
ParomomycinL. donovanipronosteady3 mth3[96]
PentostamL. donovanipronosteadyn.s.26[97]
Primaquin, pentamidine,
terbinafine, chloroquine
L. majorpronosteadyn.s.2.0–4.4[98]
Pyrimidine analogsL. mex., L. majorpronosteady12 mth1–>3500[99]
SinefuginL. infantumpronosteadyn.s.n.s.[95]
Sodium arseniteL. mex., L. amazon.pronosteady>1 mth12[89]
SbIIIL. majorpronoshockn.s.30[90]
Table 4. Risk group and biosafety level categorization of T. brucei, T. cruzi and L. donovani (in an infected insect vector, all human-pathogenic trypanosomatids are classified as biosafety level 3).
Table 4. Risk group and biosafety level categorization of T. brucei, T. cruzi and L. donovani (in an infected insect vector, all human-pathogenic trypanosomatids are classified as biosafety level 3).
CountriesT. bruceiT. cruziL. donovani
USA222
AU/NZ22n.s.
EU2 (Tbb), 3 * (Tbr)33 *
UK2 (Tbb), 3 * (Tbr)33 *
CH232
* Limited danger of transmission; usually not transmitted through the respiratory tract (Risk Group Database of the American Biological Safety Association, https://my.absa.org/Riskgroups, accessed on 19 July 2021).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Beilstein, S.; El Phil, R.; Sahraoui, S.S.; Scapozza, L.; Kaiser, M.; Mäser, P. Laboratory Selection of Trypanosomatid Pathogens for Drug Resistance. Pharmaceuticals 2022, 15, 135. https://doi.org/10.3390/ph15020135

AMA Style

Beilstein S, El Phil R, Sahraoui SS, Scapozza L, Kaiser M, Mäser P. Laboratory Selection of Trypanosomatid Pathogens for Drug Resistance. Pharmaceuticals. 2022; 15(2):135. https://doi.org/10.3390/ph15020135

Chicago/Turabian Style

Beilstein, Sabina, Radhia El Phil, Suzanne Sherihan Sahraoui, Leonardo Scapozza, Marcel Kaiser, and Pascal Mäser. 2022. "Laboratory Selection of Trypanosomatid Pathogens for Drug Resistance" Pharmaceuticals 15, no. 2: 135. https://doi.org/10.3390/ph15020135

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop