Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Phenotypic diversity and drug susceptibility of Trypanosoma cruzi TcV clinical isolates

  • Luz P. Quebrada Palacio,

    Roles Data curation, Formal analysis, Investigation, Methodology, Validation, Visualization, Writing – original draft, Writing – review & editing

    Affiliations Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina, Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina

  • Mariela N. González,

    Roles Data curation, Investigation, Methodology, Validation, Visualization, Writing – review & editing

    Current address: Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil

    Affiliation Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina

  • Yolanda Hernandez-Vasquez,

    Roles Investigation, Methodology, Validation, Visualization, Writing – review & editing

    Affiliation Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina

  • Alina E. Perrone,

    Roles Investigation, Methodology, Validation, Writing – review & editing

    Affiliation Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina

  • Adriana Parodi-Talice,

    Roles Investigation, Methodology, Supervision, Validation, Writing – review & editing

    Affiliation Unidad de Biología Molecular, Institut Pasteur de Montevideo, Sección Genética, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay

  • Jacqueline Bua,

    Roles Formal analysis, Funding acquisition, Investigation, Methodology, Resources, Supervision, Validation, Writing – review & editing

    Affiliations Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina, Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina

  • Miriam Postan

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing

    miriampostan@yahoo.com

    Affiliations Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina, Departamento de Investigación, Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina

Abstract

Trypanosoma cruzi is a genetically heterogeneous group of organisms that cause Chagas disease. It has been long suspected that the clinical outcome of the disease and response to therapeutic agents are, at least in part, related to the genetic characteristics of the parasite. Herein, we sought to validate the significance of the genotype of T. cruzi isolates recovered from patients with different clinical forms of Chagas disease living in Argentina on their biological behaviour and susceptibility to drugs. Genotype identification of the newly established isolates confirmed the reported predominance of TcV, with a minor frequency of TcI. Epimastigote sensitivity assays demonstrated marked dissimilar responses to benznidazole, nifurtimox, pentamidine and dihydroartemisinin in vitro. Two TcV isolates exhibiting divergent response to benznidazole in epimastigote assays were further tested for the expression of anti-oxidant proteins. Benznidazole-resistant BOL-FC10A epimastigotes had decreased expression of Old Yellow Enzyme and cytosolic superoxide dismutase, and overexpression of mitochondrial superoxide dismutase and tryparedoxin- 1, compared to benznidazole-susceptible AR-SE23C parasites. Drug sensitivity assays on intracellular amastigotes and trypomastigotes reproduced the higher susceptibility of AR-SE23C over BOL-FC10A parasites to benznidazole observed in epimastigotes assays. However, the susceptibility/resistance profile of amastigotes and trypomastigotes to nifurtimox, pentamidine and dihydroartemisinin varied markedly with respect to that of epimastigotes. C3H/He mice infected with AR-SE23C trypomastigotes had higher levels of parasitemia and mortality rate during the acute phase of infection compared to mice infected with BOL-FC10A trypomastigotes. Treatment of infected mice with benznidazole or nifurtimox was efficient to reduce patent parasitemia induced by either isolate. Nevertheless, qPCR performed at 70 dpi revealed parasite DNA in the blood of mice infected with AR-SE23C but not in BOL-FC10A infected mice. These results demonstrate high level of intra-type diversity which may represent an important obstacle for the testing of chemotherapeutic agents.

Introduction

It is estimated that over 7–8 million people are currently infected with Trypanosoma cruzi, the causative agent of Chagas disease [1]. Infection by this parasite results in a broad spectrum of clinical outcomes, ranging from a mild, self-resolving acute illness with low mortality rate to a potentially life-threatening cardiomyopathy, digestive and/or neurological abnormalities in up to 30% of chronically infected individuals. Chagas disease causes approximately 12,000 deaths per year [2]. Current therapy against T. cruzi relies on the nitroimidazole benznidazole (BZ) and the nitrofuran nifurtimox (NX), which are efficient to control parasitism during the acute phase of infection; however, studies on their effectiveness in the chronic phase have yielded inconsistent results [3,4].

T. cruzi is a genetically heterogeneous species of protozoan parasites transmitted by triatomine bugs distributed throughout the Americas. The large variety of host and vector species involved in the natural transmission of trypanosomes and their circulation in different eco-epidemiological settings have been held responsible for the generation of structurally different populations of parasites [5]. Following the introduction of a consensus nomenclature system for T. cruzi, namely discrete typing units (DTUs I-VI) [6,7], numerous studies have aimed to characterize the geographical distribution of T. cruzi lineages in humans and other mammals as well as their association with clinical manifestations of Chagas disease [811]. A substantial amount of experimental data relating the genotype of the parasite with biological and biochemical parameters such as parasitemia levels, tissue tropism, immune response and susceptibility to drugs have also become available, mostly for stocks collected decades ago from different sources and countries [1217]. In this study, we sought to assess the genotype of clinically relevant T. cruzi isolates obtained from infected humans of Argentina and evaluate their biological characteristics and susceptibility to conventional and investigational drugs. The results of parasite DTU identification analysis reproduced the predominant circulation of TcV reported by studies genotyping whole blood, with a minor frequency of TcI [11]. By testing drug susceptibility in vitro, we succeeded in documenting high intra-type diversity among TcV isolates, with some of them being resistant to the compounds tested. Our observations of large disparities in drug susceptibility responses are not supported by the current DTU phylogenetics classification and highlight the importance of investigating intra-type diversity in eco-epidemiological, clinical and pharmacological studies of T. cruzi.

Results

In this study, we first established a collection of T. cruzi isolates with parasites recovered from the peripheral blood of 5 chronically infected adult Chagas disease patients and 2 acutely infected children living in Argentina (2 male and 5 female). The geographic origin, age and clinical status of the subjects from whom the parasite isolates were derived are summarized in Table 1. Amplification of the non-transcribed spacer of the mini-exon genes in epimastigotes generated products of 350 bp (TcI) in 1/7 isolates (14.28%) and 300 bp (TcII, TcV and TcVI) in 6/7 (85.71%) [18,19]. Amplification of 24Sα rRNA was used to resolve the genotype of the six isolates with 300 bp amplicons in the first PCR as TcV (Table 1 and Fig 1). The occurrence of TcV isolates originated in acute and chronic infections of different severities challenges the notion that clinical outcome of Chagas disease is related to lineage-specific parasites [20].

thumbnail
Table 1. Characteristics of the Chagas disease patients with Trypanosoma cruzi positive hemoculture and parasite genotype identification.

https://doi.org/10.1371/journal.pone.0203462.t001

thumbnail
Fig 1. Genotyping of Trypanosoma cruzi isolates obtained from Chagas disease patients.

(A) PCR amplification products of the non-transcribed spacer of the mini-exon genes with primers TC, TC1 and TC2 that identify TcI (350 bp), TcII, TcV and TcVI (300 bp) and TcIII and TcIV (no amplification). (B) PCR amplification products of the D7 divergent domain of S24α rRNA with primers D71 and D72 that identify TcI, TcIII and TcV (110 bp), TcIV (120, 125 or 130 bp), TcII and TcVI (125bp). Trypanosoma cruzi strains Colombiana, CL Brener, Y, and CanIII were included as genotyping controls.

https://doi.org/10.1371/journal.pone.0203462.g001

Then, we assessed the behavior of the clinical isolates in axenic cultures. A typical exponential curve that reached the stationary phase at 8–12 days of culture was obtained for TcV isolates; the growth curve of TcI BOL-FC5C isolate was atypical in that the exponential growth phase was followed by a rapid decline in parasite density (Fig 2). The estimated doubling time of TcI BOL-FC5C (90.85 h) was significantly higher compared to that of TcV isolates (range 35.56–52.34 h; p<0.05); no significant differences in doubling time between TcV isolates were observed. The density of parasites attained by TcI BOL-FC5C cultures at the end of the exponential phase was significantly lower than that of TcV isolates cultures (p<0.05). There were also significant differences in maximum parasite density among TcV cultures (p<0.05). No correlation between doubling time and maximum parasite density was found.

thumbnail
Fig 2. Growth curves of Trypanosoma cruzi isolates recovered from humans.

Epimastigote cultures were set up in LIT media at initial concentration of 106 epimastigotes/mL and incubated at 28°C. Parasite density was estimated daily with a Neubauer’s chamber during 3 weeks.

https://doi.org/10.1371/journal.pone.0203462.g002

Afterward, we tested the sensitivity of epimastigotes to conventional drugs BZ and NX, and investigational compounds pentamidine (PENT) and dihidroartemisinin (DHA). The reference strain TcI Sylvio-X10/4, originated from a human infection, was included as susceptibility control to BZ [21] and for comparison with our newly established TcI isolate. Epimastigote sensitivity assays revealed remarkable dissimilar intra-DTU V and intra-DTU I susceptibility to each drug (p<0.05; Table 2 and S1 Fig). To establish the susceptibility/resistance phenotype, we applied the hit selection criteria for Chagas disease proposed by Katsuno et al (IC50< 10 μM) [22] and found that AR-FC195205 and BOL-FC10A epimastigotes were resistant to BZ; these isolates were also resistant to NX or PENT, respectively. None of the isolates was found to be susceptible to DHA in epimastigote sensitivity assays.

thumbnail
Table 2. Susceptibility of Trypanosoma cruzi epimastigotes to conventional chemotherapeutic agents and investigational compounds.

https://doi.org/10.1371/journal.pone.0203462.t002

To identify possible morphological changes induced by the compounds on epimastigotes, two TcV isolates derived from chronically infected adult patients exhibiting divergent sensitivity to BZ in epimastigote assays (AR-SE23C susceptible and BOL-FC10A resistant) were treated with the respective IC50 doses of each compound during 72 h at 28°C and evaluated at TEM (Fig 3). The most frequently observed changes include swelling of the mitochondrion with membrane damage and kDNA degradation, disruption of the nuclear membrane with disorganization of the chromatin, abnormal-shape reservosomes and acidocalcisomes, vacuole formation and large vesicles in the matrix, common to both isolates and the four compounds tested. Enlargement of Golgi cisternae and rare myelin-like structures were observed in parasites treated with BZ, NX and PENT, and autophagolysosomes in BZ and PENT- treated parasites. Rare abnormal kinetoplastic DNA replication was detected in PENT-treated AR-SE23C epimastigotes; spherical epimastigotes with rare swelling of the mitochondrion were observed in parasites treated with DHA.

thumbnail
Fig 3. Ultrastructural changes in epimastigotes treated with conventional and investigational anti- Trypanosoma cruzi compounds.

AR-SE23C and BOL-FC10A epimastigote cultures were incubated with IC50 doses for each compound and isolate during 72 h at 28°C; untreated epimastigotes were included as controls. Note the swelling of the mitochondrion with kDNA degradation (white arrow), myelin-like structures (arrowhead), disorganization of nuclear chromatin and nuclear membrane disruption (black arrow), the irregular replication of kDNA (curved arrow), disorganization of Golgi complex cisternae (unfilled black arrow) and autophagosomal structures (short white arrow). Bars represent 2.5 μm.

https://doi.org/10.1371/journal.pone.0203462.g003

We also measured the expression of enzymes involved in oxidant detoxification of T. cruzi in BZ-susceptible AR-SE23C and BZ-resistant BOL-FC10A epimastigotes. The expression of TcOYE and SODB, reportedly associated with susceptibility to BZ [23,24], were 1.25 and 1.36 -fold increased in AR-SE23C parasites, respectively, compared to that of BOL-FC10A parasites. In contrast, the expression of SODA and TXN 1, associated with resistance to BZ [12,25] were 2.23 -fold and 6.84 -fold higher in BOL-FC10A parasites, respectively, compared to AR-SE23C parasites (Fig 4; S2 Fig). These results support a possible relationship between drug susceptibility/resistance phenotype and T. cruzi epimastigote anti-oxidant protein expression profile.

thumbnail
Fig 4. Expression of anti-oxidant proteins in Trypanosoma cruzi.

(A) Western blotting for T. cruzi SODA, SODB, TXN 1 and TcOYE in BZ-susceptible AR-SE23C and BZ-resistant BOL-FC10A epimastigotes. (B) Densitometric analysis; bars represent the relative intensity values estimated by densitometry using Scion Imaging Software; representative assay of 2 independent experiments.

https://doi.org/10.1371/journal.pone.0203462.g004

We next tested the in vitro susceptibility/resistance profile of AR-SE23C and BOL-FC10A intracellular forms to the compounds. Drug sensitivity assays using infected peritoneal mouse macrophages (PMØ) cultures showed that the IC50 of BZ for AR-SE23C amastigotes was significantly lower than that for BOL-FC10A amastigotes (p<0.05; Table 3). Conversely, the IC50 of NX and PENT for AR-SE23C amastigotes were significantly higher compared to that of BOL-FC10A (p<0.05); no differences between IC50 values of DHA for the two isolates were found. Considering the Katsuno hit criteria [22], AR-SE23C amastigotes were susceptible to BZ, NX and PENT while BOL-FC10A amastigotes were resistant to BZ; amastigote forms of both isolates were resistant to DHA.

thumbnail
Table 3. Susceptibility of Trypanosoma cruzi AR-SE23C and BOL-FC10A amastigote and trypomastigote forms to conventional and investigational compounds.

https://doi.org/10.1371/journal.pone.0203462.t003

Cytotoxicity assays performed on uninfected PMØ cultures showed that the concentrations of BZ, NX, DHA and PENT required to reduce 50% of viable cells (CC50) were 4.84 mM (1.26 mg/mL), 4.84 mM (1.39 mg/mL), 3.30 mM (0.94 mg/mL) and 0.14 mM (0.087mg/mL), respectively. While the Selectivity Index (SI) of BZ, NX and DHA resulted >50 for AR-SE23C and BOL-FC10A amastigotes, the SI of PENT was >50 for BOL-FC10A but <50 for AR-SE23C amastigotes (SI = 35.6).

Drug sensitivity assays performed on trypomastigotes released from Vero cell cultures showed significantly higher IC50 values of BZ, NX, PENT and DHA against BOL-FC10A compared to that for AR-SE23C trypomastigotes (Table 3). While IC50 values against AR-SE23C trypomastigotes were below 10μM, IC50s against BOL-FC10A were all above that concentration. Thus, AR-SE23C trypomastigotes were considered susceptible and BOL-FC10A trypomastigotes resistant to the compounds. The four compounds had a SI >50 against AR-SE23C trypomastigotes, as well as BZ and NX against BOL-FC10A trypomastigotes; the SI of PENT and DHA against BOL-FC10A trypomastigotes was much lower (SI = 8.18 and SI = 16.09, respectively).

In order to determine the virulence of TcV BZ-susceptible AR-SE23C and BZ-resistant BOL-FC10A isolates in vivo, groups of C3H/He mice were infected with 101 to 106 Vero cell culture-derived trypomastigotes. All mice infected with AR-SE23C had variable levels of parasitemia (Fig 5A). The size of the inoculum correlated with the number of circulating parasites at the peak of parasitemia (r = 0.9429, P = 0.0167; Spearman correlation test), and higher inoculums resulted in earlier parasitemia peaks (r = -0.8827, P = 0.0333; Spearman correlation test). The estimated lethal dose 50 (LD50) of AR-SE23C was 102 parasites (Fig 5B). Circulating parasites were also detected in mice infected with 104 or 106 BOL-FC10A trypomastigotes (Fig 5C), but the area under curve (AUC) and the number of parasites at the peak of parasitemia were significantly lower than that of mice infected with the corresponding AR-SE23C inocula (P = 0.0357 and P = 0.0218, respectively; Fig 5D). No deaths were recorded in the groups of mice infected with BOL-FC10A trypomastigotes. These data indicates that isolate BOL-FC10A is significantly less virulent for C3H/He mice than isolate AR-SE23C, reaffirming the notion of biological intra-type V diversity of T. cruzi.

thumbnail
Fig 5. Course of infection in mice inoculated with Trypanosoma cruzi AR-SE23C and BOL-FC10A trypomastigotes.

(A) Parasitemia levels in C3H/He mice inoculated ip with 101 to 106 Vero cell culture-derived AR-SE23C trypomastigotes. (B) Mortality was recorded daily and analyzed using log-rank (Mantel Cox) test; P = 0.0056. (C) Parasitemia levels in mice infected with 104 and 106 Vero cell culture-derived BOL-FC10A trypomastigotes. (D) Graphical representation of parasitemia AUC in mice infected with 104 or 106 AR-SE23C or 106 BOL-FC10A trypomastigotes. ***p<0.001; t test.

https://doi.org/10.1371/journal.pone.0203462.g005

To investigate the effect of drug treatment in vivo, mice were infected with 102 AR-SE23C or 106 BOL-FC10A trypomastigotes and treated with BZ, NX or 4 PENT, starting on the day circulating parasites were first detected in the blood. BZ and NX, but not PENT prevented the increase of patent parasitemia irrespective the infecting parasite (Fig 6A and 6B); nevertheless, PENT was effective to reduce AUC (Fig 6C) and the number of blood parasites at the peak of parasitemia compared to that of non-treated infected controls (p<0.05). The data on the proportion of mice with positive T. cruzi qPCR and DNA parasite load in blood samples of mice at 70 dpi are shown in Table 4. Notably, the levels of parasite equivalents in non-treated BOL-FC10A-infected controls were significantly higher than that of non-treated AR-SE23C-infected controls with positive qPCR (p<0.05). Treatment of AR-SE23C-infected mice with PENT, but not BZ and NX significantly reduced the proportion of animals with positive qPCR; yet, the three drugs prevented mortality of mice infected with this isolate (Fig 6D). Conversely, BZ and NX but not PENT were effective to reduce the proportion of mice with positive qPCR in BOL-FC10A-infected groups.

thumbnail
Fig 6. Effect of trypanocidal drugs on the infection of mice with Trypanosoma cruzi.

Mice were inoculated with 102 AR-SE23C or 106 BOL-FC10A trypomastigotes and treated with BZ (40 doses of 100 mg/kg/d), NX (40 doses of 100 mg/kg/d) or PENT (20 doses of 4 mg/kg/d). Parasitemia curves in mice infected with AR-SE23C (A) and BOL-FC10A (B). (C) Parasitemia AUC in infected mice treated with PENT. **p<0.01 vs. non-treated infected controls; t-test. (D) Effect of treatment on survival rate of mice infected with AR-SE23C; **p<0.01 vs. non-treated infected controls; Mantel-Cox test.

https://doi.org/10.1371/journal.pone.0203462.g006

thumbnail
Table 4. Trypanosoma cruzi DNA in blood of mice infected with AR-SE23C or BOL FC10A parasites and treated with benznidazole, nifurtimox or pentamidine.

https://doi.org/10.1371/journal.pone.0203462.t004

Discussion

In nature, T. cruzi strains consist of a mixture of parasite subpopulations subjected to selective pressure by different vectors and hosts [20]. The uncovering of biochemical and molecular markers that identify distinct subpopulations within T. cruzi has led to a great advance in the understanding of parasite population dynamics and epidemiology of the infection [2630]. TcV is the most frequent genotype reported in infected humans of Argentina, followed by TcVI and TcI [11,31]. In the present study we have established 7 new human-derived T. cruzi isolates, the majority of which belong to TcV genotype with a minor representation of TcI, suggestive of them being somehow representative of the overall population currently circulating in humans of Argentina. By studying the behaviour of the parasites and performing comprehensive drug sensitivity testing, we were able to expose substantial intra-DTU phenotypic diversity within TcV, similar to that described by Mejia et al (and confirmed here) for TcI isolates [32]. In order to maintain the population as close as possible to the natural condition, all assays were performed with parasites obtained from the first 5 passages in vitro [33,34]. To favour direct contact between parasites and the drug, we established the parasite drug resistance profile in axenic culture assays. With the purpose of reducing the risk of natural parasite selection by trypanocidal drugs, we enrolled chronically and acutely infected donors with no prior anti-T.cruzi treatment. Nevertheless, a few of our isolates displayed a resistant phenotype to BZ and NX in drug sensitivity assays. It is possible that these parasites have been exposed to these drugs during circulation in domestic cycle, prior infection of the patients from whom them were retrieved.

The partial effect exerted by the conventional drugs prompted us to extend the study to investigational compounds with anti-T.cruzi action PENT and DHA, selected on the basis of their different modes of action. The mechanism of action of BZ and NX involves the reduction of the nitro groups to amino groups of the compounds by the action of T. cruzi nitroreductases, with the formation of toxic radical intermediates and electrophilic metabolites [35]. The activity of BZ against T. cruzi was proposed to be also mediated by reduced metabolites binding covalently to macromolecules such as lipids, DNA and proteins [36]. Instead, the reduction of NX leading to formation of an unsaturated open-chain nitrile was proposed as an important mechanism for its trypanocidal action [37]. Pentamidine is a synthetic aromatic diamidine widely employed for the treatment of African trypanosomiasis, leishmaniasis and infection by Pneumocystis carinii [38,39]. Trypanocidal mechanisms of diamidines include DNA binding, alteration of polyamine transport, inhibition of peptidase activity and interference with normal topoisomerase II. T. cruzi lacks the enzymes l-arginine decarboxylase and ornithine decarboxylase, required to synthesize putrescine de novo, and incorporates the polyamine from the medium through the polyamine transporter TcPAT12, which is blocked by pentamidine [40]. Artemisinin, a sesquiterpene trioxan lactone derived from Artemisia annua, and its derivatives are widely used against P. falciparum and chloroquine-resistant P. vivax. Anti-malarial action of artemisinins involves the generation of free radicals by cleavage of endoperoxide bonds through the interaction with heme iron, which alter several biochemical pathways within the parasites including the sarco-endoplasmic reticulum PfATPase6 calcium pump (SERCA) [41]. Several membrane-associated calcium ATPases have been described in T. cruzi and suggested to be responsible, at least in part, for the inhibitory effect of artemisinins on these parasites [42]. In agreement with the results reported by other authors [42], a potent action of PENT against T. cruzi epimastigotes was observed in our axenic culture assays. However, epimastigote sensitivity assays to DHA revealed that none of the human-derived isolates was susceptible to the compound. Nevertheless, AR-SE23C trypomastigotes resulted susceptible to DHA.

The comparison of the BZ susceptibility/resistance profile between amastigotes and trypomastigotes of the selected isolates reproduced that of epimastigotes (that is, AR-SE23C parasites being more susceptible than BOL-FC10A parasites). Noteworthy, BOL-FC10A epimastigotes and amastigotes were resistant to BZ but susceptible to NX. The higher activity of NX against intracellular amastigotes compared to BZ was previously noted in cell cultures infected with other strains of T. cruzi [21,43], and suggested to be related to the different modes of action of the two drugs. The pattern of drug sensitivity in our mouse model of infection did not completely translate to that of the parasites in vitro, adding further complexity to the development of effective treatments against T. cruzi. Studies on the susceptibility of T. cruzi to drugs in vivo and in vitro carried out by different laboratories have yielded inconsistent results. An example of this is Colombiana strain, which was reported resistant after 20 doses of 100 mg/kg/d BZ in vivo by Filardi and Brener [44], but susceptible as intracellular amastigotes in vitro [45]. On the other hand, Neal and van Bueren describe that in vitro drug sensitivity assays on epimastigotes and intracellular amastigotes did not distinguish between strains of T. cruzi which were responsive and non-responsive in mice, attributing this phenomenon to the host immune response against T. cruzi [43]. Conversely, other authors have shown that most drugs that are active against T. cruzi in the mouse model are also active against epimastigotes [46] or intracellular amastigotes in vitro [47]. Besides, in vitro sensitivity assays to BZ performed on epimastigotes isolated prior to treatment of patients did not correlate with the clinical therapeutic outcome [48]. The remarkable stage-dependent discrepancies in the sensitivity to drugs found in our study highlight the importance of analysing different developmental stages of the parasite in pharmacological studies of clinically relevant isolates. In this regard, Zingales proposed that natural resistance or sensitivity to drugs can be studied in the three developmental stages of T. cruzi (epimastigote, trypomastigote and amastigotes) [30].

Finally, we investigated also the possible association of parasite susceptibility/resistance to BZ with the expression of enzymes involved in the anti-redox machinery. T. cruzi synthesizes trypanothione and several enzymes that neutralize reactive oxygen and nitrogen species as a mechanism of defence against oxidative stress generated during the infection and the metabolism of trypanocidal drugs. As reported for other BZ-resistant T. cruzi populations, BOL-FC10A epimastigotes underexpressed the Old Yellow enzyme that catalyzes prostaglandin PGF2A synthesis [23,49] and SODB [24], and overexpressed SODA [25]. We also investigated the expression of tryparredoxin 1, known to be involved in oxidative metabolism and protein synthesis and degradation [50], and found it was markedly increased in BZ-resistant BOL-FC10A parasites. Although only 2 isolates were analyzed for anti-redox protein expression, our results support their association with the susceptibility/resistance phenotype of the parasites to BZ.

In summary, herein we demonstrate that T. cruzi phenotypic diversity levels are particularly high in infected humans of Argentina. Relatively few isolates were analyzed in the study, but the large variety of intra-type responses to drugs suggests that the diversity may be even more extensive than observed here. Such diversity may represent an important difficulty for the development of chemotherapeutic alternatives against Chagas disease. Assuming the present study is applicable to other DTUs, the analysis of such strains will be useful in identifying whether further parasite diversity exist.

Materials and methods

Ethics statement

A signed informed consent was obtained from all adult participants and mothers on behalf of their children. All the research was conducted in accordance with the Declaration of Helsinki and the Council for International Organizations of Medical Sciences (CIOMS). The study protocol was approved by the Review Board of Instituto Nacional de Parasitología “Dr. Mario Fatala Chaben”, Administración Nacional de Laboratorios e Institutos de Salud Dr. Carlos G. Malbrán (IRB00006651).

All animals received humane care and study protocols complied with the ARRIVE Guidelines for Reporting Animal Research [51]. The procedures were approved by the Institutional Committee for Care and Use of Laboratory Animals (CICUAL), University of Buenos Aires, School of Medicine, Secretary of Science and Technology, Buenos Aires, Argentina, Resolution No. 704/2013.

Patients

Chagas disease patients attending the Clinical Facilities of Instituto Nacional de Parasitología “Dr. Mario Fatala Chaben”, Buenos Aires, Argentina and individuals living in endemic areas of Province Santiago del Estero, Argentina, who tested positive for T. cruzi by conventional serology and had not taken anti- T. cruzi drugs were invited to participate in the study. Infected individuals with cancer, HIV infection, syphilis, diabetes or autoimmune disorders were excluded from this study. A total of 36 subjects (mean age ± SD = 46.25 ± 12.49 years; 16 males and 20 females) were recruited during the period November 2009—November 2015. The medical history and personal information such as the country of birth and current residency were recorded. The clinical status of participants was assessed by physical examination and clinical testing including electrocardiogram, echocardiography and chest radiography; 25 patients were asymptomatic, 9 patients had ECG abnormalities associated to Chagas heart disease and 2 patients had chronic Chagas dilated cardiomyopathy.

Parasite isolation

Approximately 15 mL blood were drawn from T.cruzi- infected patients by venipuncture into dry tubes (Vacutainer; BD Biosciences, Franklin Lakes, NJ, USA) and distributed in glass tubes containing biphasic media (blood-agar with liver infusion tryptose (LIT) as overlay). Samples were cultured at 28°C during 3 months and evaluated monthly for the presence of parasites at a phase microscope (Carl Zeiss, Axiostar plus, Gottingen, Germany). Supernatants from positive hemocultures were transferred to 15 mL culture tubes containing LIT media supplemented with 20 μg/mL haemin, 10% heat-inactivated foetal bovine serum (FBS), 100 U/mL penicillin, and 100 μg/mL streptomycin (complete LIT). Two additional T. cruzi isolates obtained by hemoculture of children with congenital acute infection during the same period were incorporated in this study [52]. Parasites obtained from the first 5 passages in vitro were used in all assays.

DTU identification

DNA was extracted from 108 axenic culture-derived epimastigotes with Accuprep Genomic DNA extraction kit (Bioneer Corporation, Daejeon, Korea) and used for amplification of the non- transcribed spacer of the miniexon (mini-exon genes) and the D7 divergent domain of the 24Sα rRNA gene [18]. Amplification of mini-exon with TC, TC1 and TC2 primers generate products of 300 bp (TcII, TcV and TcVI) and 350 bp (TcI); amplification of the 24Sα rRNA with D71 and D72 primers generates products of 110 (TcI, TcIII, TcV), 125 (TcII and TcVI) and 120–130 bp (TcIV). Reference strains Sylvio X10/4, Y, M5631, CanIII, Mncl2 and CL Brener were used as TcI-TcV controls.

Epimastigotes drug sensitivity testing

Epimastigote cultures were initiated at a concentration of 106 epimastigotes/mL in complete LIT and exposed to 0.1–80 μg/mL dilutions of BZ, NX, pentamidine isethionate (PENT) or dihydroartemisinin (DHA) in 96-well-plates and incubated at 28°C during 72 h. All compounds were obtained from Sigma-Aldrich (Saint Louis, MO, USA). A stock solution was prepared for each compound in dimethylsulphoxide and diluted in complete LIT media immediately prior to use in the assays; the final concentration of dimethylsulphoxide did not exceed 0.2% (v/v). At the end of each experiment, motile parasites were quantified in a Neubauer’s chamber at a phase microscope. T. cruzi Sylvio X10/4 epimastigotes were included as susceptibility controls [21]. IC50 values were estimated by means of nonlinear dose-response curve analysis using GraphPad Prism 5.0 (GraphPad Software, Inc., La Jolla, CA, USA).

Transmission electron microscopy

Epimastigotes were washed with PBS, fixed in 2.5% glutaraldehyde and 4% paraformaldehyde in 0.1 M sodium phosphate buffer pH 7.2 and embedded in 2% agarose. Samples were post-fixed in 1% osmium tetroxide—1% potassium ferrocyanide in 100 mM sodium cacodylate buffer at room temperature during 30 minutes, dehydrated in acetone and embedded in Epoxi resin. Ultrathin sections were obtained with an ultramicrotome (Reichert-Jung Ultracut E, Wien, Austria), stained with 2% uranyl acetate and lead citrate, and examined at a transmission electron microscope (Carl Zeiss 109T, Oberkochen, Germany).

Western blot

Epimastigotes were lysed using 7 M urea, 2 M thiourea, 4% CHAPS detergent, 40 mM Tris, 60 mM 1,4-Dithiothreitol (DTT) (Sigma-Aldrich, Saint Louis, MO, USA), containing the Complete Mini Protease Inhibitor Cocktail (Roche Applied Science, Indianapolis, USA) and centrifuged at 13.000×g for 30 min prior to determination of protein content by the Bradford method. Proteins were separated on 12% SDS polyacrylamide gels (20 μg/well) and transferred onto nitrocellulose membranes. To assure equal protein load in each lane, membranes were stained with Ponceau S. Blots were blocked with skimmed milk powder (5% w/v) in Tris-buffer saline containing 0.02% Tween 20 (TBS-Tween 20) for 3 h. The nitrocellulose membrane was incubated with rabbit polyclonal antibodies anti- mitochondrial superoxide dismutase (SODA; 1:2000), anti- cytosolic superoxide dismutase (SODB; 1:3000), anti- tryparedoxin- 1 (TXN 1; 1:1000) and anti- Old Yellow Enzyme (TcOYE; 1:30000), washed with TBS-Tween 20 and incubated with HRP-conjugated goat-antirabbit IgG diluted 1:2000. Signal was developed using the Chemiluminescence Detection Kit (Thermo Fisher, Waltham, MA, USA). Polyclonal antibodies against T. cruzi proteins SODA, SODB, and TXN 1 were produced as described [50,53] and TcOYE was a kind gift of Dr. Carlos Robello (Institut Pasteur, Montevideo, Uruguay).

Amastigote drug sensitivity testing

In preparation for this study, we first tested the infectivity of Vero cell-derived trypomastigotes of selected T. cruzi isolates for bovine embryo skin and muscle cells (BESM); the poor infectivity of one of the isolates for these cells prevented us from using them for the intracellular multiplication assays (S3 Fig). Then, we tested the infectivity of the parasites for freshly isolated mouse peritoneal macrophages (PMØ). Macrophages were collected from the peritoneal cavity of C3H/HeN mice by lavage with 5 mL of ice-cold RPMI medium, and suspended in 10% FBS-RPMI. Macrophages were seeded onto coverslips (5x105 cells/cover) in 24 well-plates and cultured at 37°C in a 5% CO2 incubator during 24 h. Non-adherent cells were removed by washing with RPMI and the cultures infected with Vero cell-derived AR-SE23C or BOL-FC10A trypomastigotes at a ratio of 1:1 during 16 h. After removal of non-internalized parasites, infected cultures were exposed to serial dilutions (0.01–100 μg/mL) of BZ, NX, PENT and DHA in triplicate wells during 72 h prior fixation with methanol and staining with Giemsa. The number of infected cells and intracellular amastigotes/cell was quantified in microphotographs of randomly selected 200x microscopic fields of Giemsa-stained smears (Leica CTR Mic, Germany), using Image Tool software (http://compdent.uthscsa.edu/dig/itdesc.html). A total of 600 cells (200 cells per cover) were analyzed for each experimental condition in 2 independent experiments. IC50 values were estimated by means of nonlinear dose-response curves analysis using GraphPad Prism 5.0 Software.

The effect of BZ, NX, PENT and DHA on the viability of peritoneal macrophages was measured in uninfected cell cultures using the resazurin method (Sigma-Aldrich, St. Louis, MO). Briefly, macrophages were plated in 96 well-plates (5×105 cells/well) and incubated with 0.001–5000 μg/mL dilutions of BZ, NX, PENT or DHA at a final volume of 100 μL/well during 72 h at 37 °C in a 5% CO2 incubator. Then, 10 μL of resazurin solution (0.01% in PBS) was added to each well and returned to the incubator for additional 4 h prior to recording of absorbance at 578 and 630 nm. Selectivity index of the compounds (SI) was expressed as the ratio (CC50 for macrophages/IC50 for T. cruzi parasites).

Trypomastigotes drug sensitivity testing

For drug sensitivity assays, Vero cell-derived AR-SE23C and BOL-FC10A trypomastigotes (1x106/mL) were incubated in 10% SFB-RPMI in the presence of serial dilutions (0.01–100 μg/mL) of BZ, NX, PENT and DHA for 24 h at 4°C; untreated parasites were used as controls. The number of motile parasites was determined in a Neubauer chamber at a phase microscope. IC50 were calculated as describe above.

Mice infection

The aim of this experiment was to establish the virulence of selected isolates in the mouse model, with the outcomes measures being the course of parasitemia and mortality. A total of 95 four-week-old female C3H/He mice, obtained from the Animal Breeding Facility of the Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”. Mice were selected randomly, housed in groups of five per cage at a room temperature of 22°C and provided food and water ad libitum. The cages were cleaned and sanitized daily. Mice were inoculated with serial numbers of AR-SE23C and BOL-FC10A Vero cell culture-derived trypomastigotes (101–106) via ip. The physical status, behavior and mortality were monitored by the researcher and recorded daily. The number of circulating parasites was determined in 5 μl of fresh blood samples obtained from the tail in a Neubauer chamber three times a week. Parasitemia AUC was estimated using GraphPad Prism 5.0. To minimize suffering and distress of experimental animals, humane endpoints consisting of weakness and paralysis of the posterior limbs and ataxia that could interfere with eating or drinking [54] were established in the infection protocol at the beginning of the experiment. These signs were used to decide whether to euthanize the animals during the study as a surrogate endpoint for mortality. For termination of animal studies, mice surviving the acute phase of infection were anesthetized by ip administration of a mixture of Ketamine (85mg/kg) and Xylazine (30mg/kg) and euthanized by cervical dislocation at 70 dpi (after the acute phase of infection usually subsides).

To evaluate drug susceptibility in vivo, mice were infected with AR-SE23C (102 parasites/mouse) or BOL-FC10A (106/ parasites/mouse) trypomastigotes as described above and treated with 100mg/kg/d BZ (Abarax, ELEA, Argentina) or NX (Lampit, BAYER, El Salvador) during 40 consecutive days by oral gavage, or 4mg/kg/d PENT (Pentamidina, RICHET, Argentina) during 20 consecutive days via intraperitoneal (5 mice/group). Sham-inoculated treated and non-treated infected mice were used as controls. Parasitemia and mortality were determined as mentioned above. Additional blood samples (150 μL/ mouse) were obtained by retro-orbital venipuncture prior euthanasia at 70 dpi for the detection of T. cruzi DNA by means of qPCR.

Real time PCR

Mouse blood samples were mixed with an equal volume of guanidine-HCl 6 M, EDTA 0.1 M, pH 8 (GEB), kept at room temperature for 3 days and then stored at 4°C until use. DNA was isolated from 200 μL of GEB using the High Pure PCR Template preparation Kit (Roche, Mannheim, Germany), according to the manufacturers’ protocol. Parasite DNA was amplified using a T. cruzi satellite DNA sequence of 140 bp flanked by the Sat Fw and Sat Rv oligonucleotides [55] and SYBR GreenER qPCR SuperMix Universal Kit with integrated uracil DNA glycosylase (UDG; Invitrogen, Life Technologies, Grand Island, NY, USA). DNA amplification was performed in an ABI 7500 thermocycler (Applied Biosystems, Carlsbad, CA, USA) in duplicates using 5 μL of extracted DNA as template (~100 ng) in a final volume of 20 μL. DNA extracted from non-infected mice and samples without DNA template were included as controls. qPCR conditions and standard parasite curve for data analysis were performed as previously described [52,56].

Statistical analysis

The results of assays were expressed as the mean ± SD derived from triplicate observation samples in at least two independent experiments and analyzed using GraphPad Prism. The Kolmogorov-Smirnov normality distribution test was applied to all data. Comparisons between groups were made using unpaired Student’s t test (equal variance) or Mann-Whitney test (unequal variance). Comparisons among multiple groups were made using ANOVA followed by Bonferroni or Kruskal-Wallis followed by Dunns. Values of p<0.05 were considered statistically significant.

Supporting information

S1 Fig. Dose-response curves of Trypanosoma cruzi epimastigotes.

Exponential phase epimastigotes grown in axenic cultures were treated with different dilutions of conventional and investigational trypanocidal compounds during 72 h. At the end of each experiment, motile parasites were quantified using a Neubauer’s chamber. Growth inhibition data represents the mean ± SD of combined results from three independent experiments, each including triplicate samples per dose. IC50 doses are listed in Results (Table 2).

https://doi.org/10.1371/journal.pone.0203462.s001

(TIF)

S2 Fig. Ponceau-S staining.

Membranes were stained with Ponceau S to confirm equal loading of epimastigotes samples shown in Fig 4. The molecular size marker (MW) is shown on the nitrocellulose membrane.

https://doi.org/10.1371/journal.pone.0203462.s002

(TIF)

S3 Fig. Trypanosoma cruzi infectivity in mammalian cell cultures.

Cultures of peritoneal macrophages and BESM cells were exposed to Vero cell-derived AR-SE23C and BOL-FC10A trypomastigotes during 16 h and cultured for additional 72 h. The number of infected cells was determined using Image Tool Software.

https://doi.org/10.1371/journal.pone.0203462.s003

(TIF)

Acknowledgments

We thank all the volunteers who provided blood samples for the study and Drs. Gabriela Carina Olivera and Bibiana Volta for their help in isolation of parasites. We highly appreciate the skilful technical assistance of Mrs. Claudia Nose and Mrs. Daniela Oliveto along with the Staff at the Diagnostic Department of the INP.

References

  1. 1. World Health Organization. Chagas disease (American trypanosomiasis) Fact sheet 340. 2016. http://www.who.int/mediacentre/factsheets/fs340/en/
  2. 2. Pan American Health Organization. Chagas disease. 2016. http://www.paho.org/hq/
  3. 3. Viotti R, Vigliano C, Lococo B, Bertocchi G, Petti M, Alvarez MG, et al. Long-term cardiac outcomes of treating chronic chagas disease with benznidazole versus no treatment: A nonrandomized trial. Ann Intern Med. 2006;144: 724–734. 144/10/724 pmid:16702588
  4. 4. Morillo CA, Marin-Neto JA, Avezum A, Sosa-Estani S, Rassi A, Rosas F, et al. Randomized Trial of Benznidazole for Chronic Chagas’ Cardiomyopathy. N Engl J Med. 2015;373: 1295–1306. pmid:26323937
  5. 5. Lambrecht FL. Biological variations in trypanosomes and their relation to the epidemiology of Chagas’ disease. Rev Inst Med Trop Sao Paulo. 1965;7: 346–52. Available: http://www.ncbi.nlm.nih.gov/pubmed/4955029 pmid:4955029
  6. 6. Zingales B, Andrade SG, Briones MRS, Campbell DA, Chiari E, Fernandes O, et al. A new consensus for Trypanosoma cruzi intraspecific nomenclature: second revision meeting recommends TcI to TcVI. Mem Inst Oswaldo Cruz. 2009;104: 1051–4. Available: http://www.ncbi.nlm.nih.gov/pubmed/20027478 pmid:20027478
  7. 7. Zingales B, Miles MA, Campbell DA, Tibayrenc M, Macedo AM, Teixeira MMG, et al. The revised Trypanosoma cruzi subspecific nomenclature: Rationale, epidemiological relevance and research applications. Infect Genet Evol. Elsevier B.V.; 2012;12: 240–253. pmid:22226704
  8. 8. Messenger L a, Miles M a, Bern C. Between a bug and a hard place: Trypanosoma cruzi genetic diversity and the clinical outcomes of Chagas disease. Expert Rev Anti Infect Ther. Informa UK, Ltd.; 2015;13: 995–1029. pmid:26162928
  9. 9. Apt W, Arribada A, Zulantay I, Saavedra M, Araya E, Solari A, et al. Trypanosoma cruzi burden, genotypes, and clinical evaluation of Chilean patients with chronic Chagas cardiopathy. Parasitol Res. 2015;114: 3007–3018. pmid:25935204
  10. 10. Del Puerto R, Nishizawa JE, Kikuchi M, Iihoshi N, Roca Y, Avilas C, et al. Lineage analysis of circulating Trypanosoma cruzi parasites and their association with clinical forms of chagas disease in Bolivia. PLoS Negl Trop Dis. 2010;4: 1–6. pmid:20502516
  11. 11. Cura CI, Lucero RH, Bisio M, Oshiro E, Formichelli LB, Burgos JM, et al. Trypanosoma cruzi Discrete Typing Units in Chagas disease patients from endemic and non-endemic regions of Argentina. Parasitology. 2012;139: 516–521. pmid:22309735
  12. 12. Zago MP, Hosakote YM, Koo S jie, Dhiman M, Piñeyro MD, Parodi-Talice A, et al. TcI isolates of Trypanosoma cruzi exploit the antioxidant network for enhanced intracellular survival in macrophages and virulence in mice. Infect Immun. 2016;84: 1842–1856. pmid:27068090
  13. 13. Ragone PG, Pérez Brandán C, Padilla AM, Monje Rumi M, Lauthier JJ, Alberti D’Amato AM, et al. Biological behavior of different Trypanosoma cruzi isolates circulating in an endemic area for Chagas disease in the Gran Chaco region of Argentina. Acta Trop. Elsevier B.V.; 2012;123: 196–201. pmid:22643298
  14. 14. Monteiro WM, Margioto Teston AP, Gruendling AP, dos Reis D, Gomes ML, Marques de Araújo S, et al. Trypanosoma cruzi I and IV Stocks from Brazilian Amazon Are Divergent in Terms of Biological and Medical Properties in Mice. PLoS Negl Trop Dis. 2013;7: 1–9. pmid:23437410
  15. 15. d Santos DM, Talvani A, da Mata Guedes PM, Machado-Coelho GLL, de Lana M, Bahia MT. Trypanosoma cruzi: Genetic diversity influences the profile of immunoglobulins during experimental infection. Exp Parasitol. Elsevier Inc.; 2009;121: 8–14. pmid:18848935
  16. 16. Moraes CB, Giardini M a, Kim H, Franco CH, Araujo-Junior AM, Schenkman S, et al. Nitroheterocyclic compounds are more efficacious than CYP51 inhibitors against Trypanosoma cruzi: implications for Chagas disease drug discovery and development. Sci Rep. 2014;4: 4703. pmid:24736467
  17. 17. Aquilino C, Rubio ML, Seco EM, Escudero L, Corvo L, Soto M, et al. Differential trypanocidal activity of novel macrolide antibiotics; correlation to genetic lineage. PLoS One. 2012;7. pmid:22859958
  18. 18. Brisse S, Verhoef J, Tibayrenc M. Characterisation of large and small subunit rRNA and mini-exon genes further supports the distinction of six Trypanosoma cruzi lineages. Int J Parasitol. 2001;31: 1218–1226. pmid:11513891
  19. 19. Lewis MD, Ma J, Yeo M, Carrasco HJ, Llewellyn MS, Miles MA. Genotyping of Trypanosoma cruzi: Systematic selection of assays allowing rapid and accurate discrimination of all known lineages. Am J Trop Med Hyg. 2009;81: 1041–1049. pmid:19996435
  20. 20. Macedo AM, Oliveira RP, Pena SDJ. Chagas disease: role of parasite genetic variation in pathogenesis. Expert Rev Mol Med. 2002;4: 1–16.
  21. 21. Luna KP, Hernández IP, Rueda CM, Zorro MM, Croft SL, Escobar P. In vitro susceptibility of Trypanosoma cruzi strains from Santander, Colombia, to hexadecylphosphocholine (miltefosine), nifurtimox and benznidazole. Biomedica. 2009;29: 448–55. pmid:20436996
  22. 22. Katsuno K, Burrows JN, Duncan K, Hooft van Huijsduijnen R, Kaneko T, Kita K, et al. Hit and lead criteria in drug discovery for infectious diseases of the developing world. Nat Rev Drug Discov. Nature Publishing Group; 2015;14: 751–8. pmid:26435527
  23. 23. Andrade HM, Murta SMF, Chapeaurouge A, Perales J, Nirdé P, Romanha AJ. Proteomic analysis of Trypanosoma cruzi resistance to benznidazole. J Proteome Res. 2008;7: 2357–2367. pmid:18435557
  24. 24. Temperton NJ, Wilkinson SR, Meyer DJ, Kelly JM. Overexpression of superoxide dismutase in Trypanosoma cruzi results in increased sensitivity to the trypanocidal agents gentian violet and benznidazole. Mol Biochem Parasitol. 1998;96: 167–76. pmid:9851615
  25. 25. Nogueira FB, Krieger MA, Nirdé P, Goldenberg S, Romanha AJ, Murta SMF. 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. pmid:17113553
  26. 26. Ready PD, Miles MA. Delimitation of Trypanosoma cruzi zymodemes by numerical taxonomy. Trans R Soc Trop Med Hyg. 1980;74: 238–42. Available: http://www.ncbi.nlm.nih.gov/pubmed/6992359 pmid:6992359
  27. 27. Brisse S, Dujardin JC, Tibayrenc M. Identification of six Trypanosoma cruzi lineages by sequence-characterised amplified region markers. Mol Biochem Parasitol. 2000;111: 95–105. pmid:11087920
  28. 28. Yeo M, Mauricio IL, Messenger LA, Lewis MD, Llewellyn MS, Acosta N, et al. Multilocus sequence typing (MLST) for lineage assignment and high resolution diversity studies in Trypanosoma cruzi. PLoS Negl Trop Dis. 2011;5: e1049. pmid:21713026
  29. 29. Tibayrenc M, Ayala FJ. The population genetics of Trypanosoma cruzi revisited in the light of the predominant clonal evolution model. Acta Trop. Elsevier B.V.; 2015;151: 156–165. pmid:26188332
  30. 30. Zingales B. Trypanosoma cruzi genetic diversity: Something new for something known about Chagas disease manifestations, serodiagnosis and drug sensitivity. Acta Trop. Elsevier B.V.; 2017; pmid:28941731
  31. 31. Monje-Rumi MM, Brandán CP, Ragone PG, Tomasini N, Lauthier JJ, Alberti D’Amato AM, et al. Trypanosoma cruzi diversity in the Gran Chaco: Mixed infections and differential host distribution of TcV and TcVI. Infect Genet Evol. 2015;29: 53–59. pmid:25445658
  32. 32. Mejia AM, Hall BS, Taylor MC, Gómez-Palacio A, Wilkinson SR, Triana-Chávez O, et al. Benznidazole-resistance in Trypanosoma cruzi is a readily acquired trait that can arise independently in a single population. J Infect Dis. 2012;206: 220–8. pmid:22551809
  33. 33. Bice DE, Zeledon R. Comparison of infectivity of strains of Trypanosoma cruzi (Chagas, 1909). J Parasitol. 1970;56: 63–7. Available: http://www.ncbi.nlm.nih.gov/pubmed/5459047 pmid:5459047
  34. 34. Urdaneta-Morales S. Trypanosoma cruzi: attenuation of virulence by culture in tissues. Ann Parasitol Hum Comp. 1983;58: 317–24. Available: http://www.ncbi.nlm.nih.gov/pubmed/6416130 pmid:6416130
  35. 35. Maya JD, Cassels BK, Iturriaga-Vásquez P, Ferreira J, Faúndez M, Galanti N, et al. Mode of action of natural and synthetic drugs against Trypanosoma cruzi and their interaction with the mammalian host. Comp Biochem Physiol—A Mol Integr Physiol. 2007;146: 601–620. pmid:16626984
  36. 36. Trochine A, Creek DJ, Faral-Tello P, Barrett MP, Robello C. Benznidazole biotransformation and multiple targets in Trypanosoma cruzi revealed by metabolomics. PLoS Negl Trop Dis. 2014;8: e2844. pmid:24853684
  37. 37. Hall BS, Bot C, Wilkinson SR. Nifurtimox activation by trypanosomal type I nitroreductases generates cytotoxic nitrile metabolites. J Biol Chem. 2011;286: 13088–95. pmid:21345801
  38. 38. Kaiser M, Mäser P, Tadoori LP, Ioset JR, Brun R, Sullivan DJ. Antiprotozoal activity profiling of approved drugs: A starting point toward drug repositioning. PLoS One. 2015;10: 1–16. pmid:26270335
  39. 39. MacIejewska D, Zabinski J, Kaźmierczak P, Rezler M, Krassowska-Świebocka B, Collins MS, et al. Analogs of pentamidine as potential anti-Pneumocystis chemotherapeutics. Eur J Med Chem. 2012;48: 164–173. pmid:22200403
  40. 40. Díaz M V., Miranda MR, Campos-Estrada C, Reigada C, Maya JD, Pereira CA, et al. Pentamidine exerts in vitro and in vivo anti Trypanosoma cruzi activity and inhibits the polyamine transport in Trypanosoma cruzi. Acta Trop. 2014;134: 1–9. pmid:24560964
  41. 41. Ho WE, Peh HY, Chan TK, Wong WSF. Artemisinins: pharmacological actions beyond anti-malarial. Pharmacol Ther. 2014;142: 126–39. pmid:24316259
  42. 42. Mishina Y V., Krishna S, Haynes RK, Meade JC. Artemisinins inhibit Trypanosoma cruzi and Trypanosoma brucei rhodesiense in vitro growth. Antimicrob Agents Chemother. 2007;51: 1852–1854. pmid:17339374
  43. 43. Neal RA, van Bueren J. Comparative studies of drug susceptibility of five strains of Trypanosoma cruzi in vivo and in vitro. Trans R Soc Trop Med Hyg. 1988;82: 709–14. https://doi.org/10.1016/0035-9203(88)90208-8 pmid:3075357
  44. 44. Filardi LS, Brener Z. Susceptibility and natural resistance of Trypanosoma cruzi strains to drugs used clinically in Chagas disease. Trans R Soc Trop Med Hyg. 1987;81: 755–9. Available: http://www.sciencedirect.com/science/article/pii/0035920387900204 pmid:3130683
  45. 45. Canavaci AMC, Bustamante JM, Padilla AM, Perez Brandan CM, Simpson LJ, Xu D, et al. In vitro and in vivo high-throughput assays for the testing of anti-Trypanosoma cruzi compounds. PLoS Negl Trop Dis. 2010;4: e740. pmid:20644616
  46. 46. Schlemper BR, Chiari E, Brener Z. Growth-inhibition drug test with Trypanosoma cruzi culture forms. J Protozool. 1977;24: 544–7. pmid:340677
  47. 47. Croft SL. In vitro screens in the experimental chemotherapy of leishmaniasis and trypanosomiasis. Parasitol Today. 1986;2: 64–9. pmid:15462773
  48. 48. Moreno M, D’ávila DA, Silva MN, Galvão LM, Macedo AM, Chiari E, et al. Trypanosoma cruzi benznidazole susceptibility in vitro does not predict the therapeutic outcome of human Chagas disease. Mem Inst Oswaldo Cruz. 2010;105: 918–24. Available: http://www.ncbi.nlm.nih.gov/pubmed/21120364 pmid:21120364
  49. 49. Kubata BK, Kabututu Z, Nozaki T, Munday CJ, Fukuzumi S, Ohkubo K, et al. A key role for old yellow enzyme in the metabolism of drugs by Trypanosoma cruzi. J Exp Med. 2002;196: 1241–1251. pmid:12417633
  50. 50. Piñeyro MD, Parodi-Talice A, Portela M, Arias DG, Guerrero SA, Robello C. Molecular characterization and interactome analysis of Trypanosoma cruzi tryparedoxin 1. J Proteomics. 2011;74: 1683–92. pmid:21539948
  51. 51. Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: The ARRIVE guidelines for reporting animal research. J Pharmacol Pharmacother. 2010;1: 94–9. pmid:21350617
  52. 52. Bua J, Volta BJ, Perrone AE, Scollo K, Velázquez EB, Ruiz AM, et al. How to Improve the Early Diagnosis of Trypanosoma cruzi Infection: Relationship between Validated Conventional Diagnosis and Quantitative DNA Amplification in Congenitally Infected Children. PLoS Negl Trop Dis. 2013;7: 12–14. pmid:24147166
  53. 53. Peloso EF, Gonçalves CC, Silva TM, Ribeiro LHG, Piñeyro MD, Robello C, et al. Tryparedoxin peroxidases and superoxide dismutases expression as well as ROS release are related to Trypanosoma cruzi epimastigotes growth phases. Arch Biochem Biophys. 2012;520: 117–22. pmid:22425758
  54. 54. Postan M, Dvorak JA, McDaniel JP. Studies of Trypanosoma cruzi clones in inbred mice. I. A comparison of the course of infection of C3H/HEN- mice with two clones isolated from a common source. Am J Trop Med Hyg. 1983;32: 497–506. Available: http://www.ncbi.nlm.nih.gov/pubmed/6407346 pmid:6407346
  55. 55. Duffy T, Bisio M, Altcheh J, Burgos JM, Diez M, Levin MJ, et al. Accurate real-time PCR strategy for monitoring bloodstream parasitic loads in chagas disease patients. PLoS Negl Trop Dis. 2009;3: e419. pmid:19381287
  56. 56. Bua J, Volta BJ, Velazquez EB, Ruiz AM, De Rissio AM, Cardoni RL. Vertical transmission of Trypanosoma cruzi infection: quantification of parasite burden in mothers and their children by parasite DNA amplification. Trans R Soc Trop Med Hyg. 2012;106: 623–8. pmid:22835758