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

Optimization of a Fluorescence-Based Assay for Large-Scale Drug Screening against Babesia and Theileria Parasites

  • Mohamed Abdo Rizk ,

    Contributed equally to this work with: Mohamed Abdo Rizk, Shimaa Abd El-Salam El-Sayed

    Affiliations National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan, Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt

  • Shimaa Abd El-Salam El-Sayed ,

    Contributed equally to this work with: Mohamed Abdo Rizk, Shimaa Abd El-Salam El-Sayed

    Affiliations National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan, Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt

  • Mohamed Alaa Terkawi,

    Affiliation National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan

  • Mohamed Ahmed Youssef,

    Affiliation Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt

  • El Said El Shirbini El Said,

    Affiliation Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt

  • Gehad Elsayed,

    Affiliation Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt

  • Sabry El-Khodery,

    Affiliation Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt

  • Maged El-Ashker,

    Affiliation Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt

  • Ahmed Elsify,

    Affiliations National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan, Department of Animal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Minoufiya University, Sadat City, Minoufiya, Egypt

  • Mosaab Omar,

    Affiliation Department of Parasitology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt

  • Akram Salama,

    Affiliation Department of Animal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Minoufiya University, Sadat City, Minoufiya, Egypt

  • Naoaki Yokoyama,

    Affiliation National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan

  • Ikuo Igarashi

    igarcpmi@obihiro.ac.jp

    Affiliation National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan

Abstract

A rapid and accurate assay for evaluating antibabesial drugs on a large scale is required for the discovery of novel chemotherapeutic agents against Babesia parasites. In the current study, we evaluated the usefulness of a fluorescence-based assay for determining the efficacies of antibabesial compounds against bovine and equine hemoparasites in in vitro cultures. Three different hematocrits (HCTs; 2.5%, 5%, and 10%) were used without daily replacement of the medium. The results of a high-throughput screening assay revealed that the best HCT was 2.5% for bovine Babesia parasites and 5% for equine Babesia and Theileria parasites. The IC50 values of diminazene aceturate obtained by fluorescence and microscopy did not differ significantly. Likewise, the IC50 values of luteolin, pyronaridine tetraphosphate, nimbolide, gedunin, and enoxacin did not differ between the two methods. In conclusion, our fluorescence-based assay uses low HCT and does not require daily replacement of culture medium, making it highly suitable for in vitro large-scale drug screening against Babesia and Theileria parasites that infect cattle and horses.

Introduction

Babesiosis is a tick-transmitted disease caused by hemoprotozoan parasites and known to bring great economic losses in the bovine and equine industries worldwide. Bovine babesiosis caused by B. bovis and B. bigemina have a considerable impact on cattle health and productivity [1]. Equine piroplasmosis, caused by T. equi [2] and B. caballi [3], is considered one of the most important protozoan diseases affecting horses, mules, and donkeys [3]. Clinical manifestations of babesiosis include fever, hemolytic anemia, hemoglobinuria, and sometimes death [1, 4]. To date, chemotherapy by imidocarb dipropionate and diminazene aceturate is the most common strategy for controlling infection in the field [5]. However, concerns about the toxicity and resistance of these drugs have emerged [5]. Therefore, there is an urgent need to develop more effective and safer antibabesial drugs.

Microscopic examination of stained blood smears is the gold standard for direct detection of parasites. However, this method is influenced by the quality of the blood smears as well as the skill and experience of experimentators and is not suitable for large-scale drug screening [6]. A new, alternative strategy that offers accuracy, simplicity, and automatic analysis is absolutely necessary [7]. To that end, a fluorescence-based technique using SYBR Green I (SGI) has been proposed as it provides more reliable results in a short time without being influenced by experimentator variations. In fact, this assay has been employed to evaluate the efficacies of antimalarials in vitro [8]. In brief, the assay relies on the high throughput screening of red blood cells to detect parasite DNA using a fluorescent spectrophotometer [8]. High-throughput screening (HTS) assay is a well-established process for drug discovery in biotech companies [9]. During the last 2 decades, there has been a dramatic increase in the number of available compounds, leading to a fundamental change in the drug discovery process applied in research units. Strikingly, HTS assay has the ability to test 10,000 to 100,000 compounds per day [9]. Therefore, this assay is one suitable approach to mass screening of compounds.

Although we have recently developed a novel fluorescence-based assay in our laboratory for drug evaluations against B. bovis [10], the assay is not suitable for large-scale screening of drugs due to the need for daily replacement of the medium, which is time consuming and requires great effort. Moreover, the previous assay has been shown to lower coefficients of variation at the maximal signal (% CVmax) and the minimal signal (% CVmin) percentages in comparison with the malaria parasites [11]. Therefore, in the present study, we optimized the assay to be suitable for large-scale screening of drugs and evaluated its usefulness in monitoring the in vitro growth of B. bovis, B. bigemina, T. equi, and B. caballi treated with diminazene aceturate, luteolin, pyronaridine tetraphosphate, nimbolide, gedunin, or enoxacin. The study revealed the promising use of our optimized fluorescence based assay for high throughput screening of compounds against in vitro growth of Babesia and Theileria parasites.

Material and Methods

Ethics statement

Experiments described in this article were conducted in accordance with the Guiding Principles for the Care and Use of Research Animals promulgated by Obihiro University of Agriculture and Veterinary Medicine, Japan. The protocol was approved by the Committee on the Ethics of Animal Experiments of the Obihiro University of Agriculture and Veterinary Medicine (Permit numbers 26–27).

Chemical reagents

SYBR Green I (SGI) nucleic acid stain (Lonza, USA; 10,000x) was stored at -20°C and thawed before use. A lysis buffer consisting of Tris (130 mM; pH 7.5), EDTA (10 mM), saponin (0.016%; W/V), and TritonX-100 (1.6%; V/V) was prepared in advance and stored at 4°C. Diminazene aceturate (Novartis, Japan), luteolin (Sigma-Aldrich, Japan), pyronaridine tetraphosphate (Sigma-Aldrich), nimbolide (BioVision, USA), gedunin (Tocris Bioscience, UK), and enoxacin (Sigma-Aldrich) were prepared as 100 mM or 50 mM stock solutions.

In vitro cultures of Babesia parasites

A Texas strain of B. bovis [12], an Argentina strain of B. bigemina [13], a U.S. Department of Agriculture (USDA) strain of B. caballi [14], and a USDA strain of T. equi (B. equi) [15] were cultivated in purified bovine or equine red blood cells (RBCs) using a microaerophilic, stationary-phase culture system [16, 17]. Briefly, Medium 199 was used for B. bovis, B. bigemina, and T. equi whereas RPMI 1640 was used for B. caballi (both from Sigma-Aldrich, Japan). Media were supplemented with 40% normal bovine serum (for bovine Babesia isolates) or 40% normal horse serum (for equine Babesia and Theileria isolates), 60 U/ml penicillin G, 60 μg/ml streptomycin, and 0.15 μg/ml amphotericin B (all three drugs from Sigma-Aldrich). Additionally, 13.6 μg of hypoxanthine (ICN Biomedicals, Inc., USA) per ml was added to T. equi culture as a vital supplement. Cultures of parasitized RBCs (pRBCs) were incubated at 37°C in an atmosphere of 5% CO2, 5% O2, and 90% N2. After 4 days of culture, the percentages of infected RBCs reached 9.6 to 11.2% for B. bovis, 5.1 to 6.7% for B. bigemina, 8.1 to 15.0% for T. equi, and 6.5 to 8.7% for B. caballi.

Assessment of SYBR Green I fluorescence linearity

A 96-well plate was used to assess the linearity between the fluorescent values and parasitemia values as determined by microscopy. Briefly, B. bovis, B. bigemina, T. equi, and B. caballi pRBCs were serially diluted with non-parasitized bovine or equine RBCs to parasitemia ranging from 0.25% to 8% in 100 uL [18]. Non-parasitized bovine or equine RBCs were used as a blank control. Bovine RBCs were prepared in M199 medium; equine RBCs were prepared in M199 medium for T. equi and RPMI 1640 medium for B. caballi in triplicate at three different hematocrit (HCT) (2.5%, 5%, and 10%). One hundred μl of a lysis buffer was mixed with a 2× SGI (10,000x) nucleic acid stain and added directly to each dilution by gentle mixing. Plates were incubated for 24 hours in a dark place at room temperature, and fluorescence values determined using a fluorescence plate reader (Fluoroskan Ascent, Thermo Labsystems, USA) at 485 nm and 518 nm excitation and emission wavelengths, respectively. Gain values were set to 100. The Parasitemia values were plotted against RFU values, after substraction of non-parasitized bovine or equine RBCs, and analyzed by linear regression.

Determination of statistical parameters for high-throughput screening (HTS) assays

B. bovis, B. bigemina, T. equi, and B. caballi pRBCs at 1% parasitemia were cultivated in 96-well plates with non-parasitized bovine or equine RBCs at three different HCT percentages (2.5%, 5%, and 10%). Culture medium was M199 or RPMI 1640 alone (positive growth, 100% growth) or supplemented with a supralethal dose (20 uM) of diminazene aceturate (negative control). Then, the cultures of pRBCs were incubated without daily replacement of the medium, and fluorescence values were determined after the fourth day as previously mentioned. Nine samples were used for each medium in triplicate.

Determination of assay quality

Statistical analyses to determine assay quality (or Z’) were performed according to [19]. Briefly, the equations for calculating the Z’ factor (Z’), signal to noise (S/N) ratio, coefficient of variation at the maximum signal (% CVmax, positive control), and coefficient of variation at the minimum signal (% CVmin, negative control) are as follows: Z’ = 1–[(3σ(+)+3σ(-)/μ(+)-μ(-)], S/N ratio = [μ(+)-μ(-)]/σ(-), % CVmax = 100x[σ(+)/μ(+)], and % CVmin = 100x[σ(-)/μ(-)], respectively, where μ(+) and σ(+) are the mean and standard deviation of pRBCs (positive control), respectively, and μ(-) and σ(-) are the mean and standard deviation of the uninfected bovine and equine RBCs (negative control), respectively.

Optimization of the fluorescence-based assay

B. bovis and T. equi pRBCs at 1% parasitemia were cultivated in 96-well plates at different HCT values (2.5%, 5%, and 10%) in M199 medium alone or with the indicated concentrations of diminazene aceturate (10, 250, 500, 1000, 5000, and 10000 nM) in 100 μl final volume. Non-parasitized RBCs were loaded into each well in triplicate and used as a blank control. RBC cultures were maintained for 4 days without replacement of the medium. On the fourth day, thin blood smears from each well were prepared and stained with Giemsa stain to be observed under the microscope to calculate parasitemia. Next, 100 μl of a lysis buffer containing 2× SGI nucleic acid stain was directly added to each well by gentle mixing. The plates were then incubated for 24 hours in a dark place at room temperature, and the fluorescence values were determined using a fluorescence plate reader as previously mentioned. Gain values were set to 100. The mean fluorescence values (after background subtraction of uninfected bovine and equine RBCs) were then plotted against the logarithm of drug concentrations. The 50% inhibitory concentration (IC50) values were calculated on the fourth day of culture using both the fluorescence-based assay and microscopic examination. In a separate experiment, two 96-well plates were used for the cultivation of B. bovis and T. equi pRBCs at 1% parasitemia at 2.5% and 5% HCTs, respectively. The pRBCs were cultivated for 4 days with replacement of the medium in triplicate wells for each concentration of the drug. On the fourth day, IC50 values of diminazene aceturate were determined as reported above. Each experiment was repeated three times.

Antibabesial drug screening by fluorescence-based assay

B. bovis, B. bigemina, T. equi, and B. caballi pRBCs were cultivated at 1% parasitemia in 96-well plates using 2.5% and 5% HCTs for bovine and equine parasites, respectively, and M199 or RPMI 1640 media alone or with the indicated concentrations of drugs, including diminazene aceturate, luteolin, pyronaridine tetraphosphate, nimbolide, gedunin, and enoxacin. The pRBCs were cultivated for 4 days without replacement of the medium in triplicate wells for each concentration of the drug. Four plates were used in quadruplicate for each drug experiment in bovine Babesia parasites and in diminazene aceturate only for equine Babesia and Theileria parasites, and growth inhibition assays [20] between 1 and 4 days post-drug addition were conducted to evaluate whether the drug-induced growth inhibition is reflected by fluorescence signals in a dose-dependent manner. With other drugs for equine Babesia and Theileria parasites, only one plate was used in each drug experiment for 50% inhibitory concentration (IC50) value calculation on the fourth day of culture after adding lysis buffer containing 2× SGI. The IC50 values were calculated on the fourth day of culture using both the fluorescence-based assay and microscopic examination. Each experiment was repeated three times.

Statistical analysis

Data analysis was performed using GraphPad Prism ver. 5 (GraphPad Software, Inc., USA) using the one-way ANOVA. The IC50 values were calculated using GraphPad Prism ver. 5. The mean IC50 values by fluorescence assay and microscopic examination for each tested drug were analyzed using an unpaired t-test with the GraphPad software available online (http://www.graphpad.com/quickcalcs/ttest1.cfm?Format=SD). P < 0.05 was considered to be statistically significant.

Results

Assessment of SYBR Green I fluorescence linearity

The correlation between the fluorescence values and microscopy values was assessed in this assay using three different HCTs (2.5%, 5%, and 10%). Correlating relative fluorescence units (RFUs) with parasitemias for bovine Babesia parasites showed significant linear relationships with R2 values of 0.9945, 0.9328, and 0.9930 for B. bovis and R2 values of 0.9842, 0.9637, and 0.9387 for B. bigemina with HCTs of 2.5%, 5%, and 10%, respectively (Fig 1A and 1B). For equine Babesia and Theileria parasites, significant linear relationships between the fluorescence and parasitemia values were only seen for 5% and 10% HCTs with R2 values of 0.9764 and 0.9946 for T. equi and R2 values of 0.9786 and 0.9315 for B. caballi (Fig 1C and 1D). In summary, strong correlations (R2 > 0.97) were obtained at 2.5% and 10% HCTs for B. bovis, 2.5% HCT for B. bigemina, 5% and 10% HCTs for T. equi, and 5% HCT for B. caballi.

thumbnail
Fig 1. Linearity assessment between relative fluorescence readings and parasitemia percentages of B. bovis, B. bigemina, T. equi, and B. caballi pRBCs with different HCTs: 2.5%, 5%, and 10%.

(A) B. bovis pRBCs. (B) B. bigemina pRBCs. (C) T. equi pRBCs. (D) B. caballi pRBCs. Each value is presented as the mean of triplicates after subtraction of the background fluorescence for non-parasitized RBCs.

https://doi.org/10.1371/journal.pone.0125276.g001

Validation of modified high-throughput screening (HTS) assays

To evaluate the quality of the assay, the Z’ factor, S/N ratio, % CVmax, and % CVmin were calculated for each of the three HCTs (2.5%, 5%, and 10%). For bovine Babesia parasites, the Z’ factors were similar for all HCTs within a range of 0.60 to 0.97 (S1 Table). On the other hand, S/N ratios showed the highest values at 2.5% HCT (S1 Table). Regarding the equine parasites, the Z' factor values were greater than 0.5 at 5% HCT but lower than 0.5 for the other two HCTs (S1 Table). In addition, S/N ratios had the highest values at 5% HCT for both parasites. These results reveal the robustness and accuracy of the fluorescence-based assay with 2.5% and 5% HCTs without daily replacement of the medium for bovine and equine parasites, respectively.

Optimization of the fluorescence-based assay

Optimization of the fluorescence-based assay required determination of the IC50 values of diminazene aceturate for B. bovis and T. equi by fluorescence- and microscopy-based methods in cultured parasites after 4 days of treatment without daily replacement of the medium at the optimal HCTs (2.5% and 5% for B. bovis and T. equi, respectively) in three separate experiments. The results obtained by the two assays at 2.5% and 5% HCTs, for B. bovis and T. equi, respectively were not significantly different (Table 1). Next, to exclude the death of parasites due to the non-daily replacement of the medium, the same experiment was applied with 2.5% HCT for B. bovis and 5% HCT for T. equi with daily replacement of the medium, and the IC50 value was calculated by the fluorescence-based method. The results revealed no significant difference between the IC50 values obtained from the experiment with daily replacement of the medium and that without daily replacement of the medium (Table 2; Fig 2A and 2B). The absolute values obtained from the fluorescence—based assay after background subtraction of uninfected bovine RBCs for both cultures of B. bovis are also reported (S4A Fig). These results revealed the usefulness of the fluorescence-based assay for mass drug screenings in in vitro cultures of bovine Babesia parasites and equine Babesia and Theileria parasites with 2.5% and 5% HCTs, respectively, without daily replacement of the medium.

thumbnail
Table 1. IC50 values of the diminazene aceturate drug evaluated for B. bovis and T. equi by fluorescence- and microscopy-based methods without daily replacement of the medium with the optimal HCTs.

https://doi.org/10.1371/journal.pone.0125276.t001

thumbnail
Table 2. IC50 values of the diminazene aceturate drug evaluated for B. bovis and T. equi with and without daily replacement of the medium using the fluorescence-based method.

https://doi.org/10.1371/journal.pone.0125276.t002

thumbnail
Fig 2. Growth inhibition of B. bovis and T. equi by diminazene aceturate estimated by the fluorescence-based method without daily replacement of the medium (black) and with daily replacement of the medium (gray).

(A) Growth inhibition of B. bovis (y-axis) and log concentration of diminazene aceturate (nM) (x-axis). (B) Growth inhibition of T. equi (y-axis) and log concentration of diminazene aceturate (nM) (x-axis). Each value is presented as the mean of three triplicate wells ± SD after subtraction of the background fluorescence for non-parasitized RBCs.

https://doi.org/10.1371/journal.pone.0125276.g002

Fluorescence-based assay for evaluation of the in vitro growth inhibitory effects

We assessed parasitemia using the fluorescence based assay in day 1 to day 4 RBC cultures infected with four hemoparasites and exposed to six drugs (Figs 3 and 4; S1 and S2 Figs). Data showed that fluorescence values were inversely correlated with drug concentrations.

thumbnail
Fig 3. Fluorescence-based monitoring of diminazene aceturate-induced growth inhibition of B. bovis, B. bigemina, T. equi, and B. caballi during 4 days of treatment.

(A) Correlation between RFUs (y-axis) and log concentration of diminazene aceturate (nM) (x-axis) on B. bovis. (B) Correlation between RFUs (y-axis) and log concentration of diminazene aceturate (nM) (x-axis) on B. bigemina. (C) Correlation between RFUs (y-axis) and log concentration of diminazene aceturate (nM) (x-axis) on T. equi. (D) Correlation between RFUs (y-axis) and log concentration of diminazene aceturate (nM) (x-axis) on B. caballi. Each value represents a mean of triplicate wells after subtraction of the background fluorescence for non-parasitized RBCs.

https://doi.org/10.1371/journal.pone.0125276.g003

thumbnail
Fig 4. Fluorescence-based monitoring of luteolin-, pyronaridine tetraphosphate-, and nimbolide-induced growth inhibition of B. bovis, B. bigemina, T. equi, and B. caballi on the fourth day of treatment.

(A) Correlation between luteolin, pyronaridine tetraphosphate, and nimbolide concentrations and RFUs on B. bovis. (B) Correlation between luteolin, pyronaridine tetraphosphate, and nimbolide concentrations and RFUs on B. bigemina. (C) Correlation between luteolin, pyronaridine tetraphosphate, and nimbolide concentrations and RFUs on T. equi. (D) Correlation between luteolin, pyronaridine tetraphosphate, and nimbolide concentrations and RFUs on B. caballi. Each value represents a mean of triplicate wells after subtraction of the background fluorescence for non-parasitized RBCs. Statistically significant differences are indicated by asterisks (*P <0.05) between the drug-treated cultures and the control cultures.

https://doi.org/10.1371/journal.pone.0125276.g004

The maximum parasitemias of all parasites in this study in the control cultures were observed on the fourth day. Therefore, the fluorescence values of the experiments were determined on the fourth day of treatment.

The IC50 values for the six drugs commonly used in the field, such as diminazene aceturate, luteolin, pyronaridine tetraphosphate, nimbolide, gedunin, and enoxacin were determined and compared to those obtained by the microscopy method. These drugs were used with the indicated concentrations: 5, 10, 100, 250, 500, 1000, 2500, 5000, 10000, and 15000 nM for diminazene aceturate; 10, 250, 500, 1000, 5000, 10000, 25000, 50000, 100000, 200000, and 400000 nM for luteolin, pyronaridine tetraphosphate, nimbolide, and gedunin; and 10, 250, 500, 1000, 5000, 10000, 25000, 50000, and 100000 nM for enoxacin. Strikingly, the IC50 values obtained from the current assay were significantly concordant with those obtained by microscopy (P > 0.05) in all tested drugs for the four parasites (Table 3; Fig 5). Additionally, the absolute values obtained from the fluorescence-based assay after background subtraction of uninfected bovine RBCs and that obtained by the microscopy method for the culture of B. bovis without daily replacement of the medium are also reported (S4B and S4C Fig). These results confirmed the successful application of our fluorescence-based assay for mass drug screenings against bovine and equine Babesia and Theileria parasites in in vitro cultures.

thumbnail
Table 3. IC50 values of antibabesial drugs evaluated for bovine Babesia and equine Babesia and Theileria parasites using fluorescence- and microscopy-based methods without daily replacement of the medium with 2.5% and 5% HCTs, respectively.

https://doi.org/10.1371/journal.pone.0125276.t003

thumbnail
Fig 5. Growth inhibition of B. bovis, B. bigemina, T. equi, and B. caballi by diminazene aceturate on the fourth day estimated by the fluorescence-based method (black) and microscopy (gray).

(A) Growth inhibition of B. bovis (y-axis) and log concentration of diminazene aceturate (nM) (x-axis). (B) Growth inhibition of B. bigemina (y-axis) and log concentration of diminazene aceturate (nM) (x-axis). (C) Growth inhibition of T. equi (y-axis) and log concentration of diminazene aceturate (nM) (x-axis). (D) Growth inhibition of B. caballi (y-axis) and log concentration of diminazene aceturate (nM) (x-axis). Each value is presented as the mean of three triplicate wells ± SD.

https://doi.org/10.1371/journal.pone.0125276.g005

The in vitro growth of B. bovis was significantly inhibited (P < 0.05) by 10, 1000, 1000, 500 and 50000 nM luteolin, pyronaridine tetraphosphate, nimbolide, gedunin, and enoxacin, respectively (Fig 4A; S1D and S1E Fig); while, B. bigemina significantly inhibited by the same drugs at 250, 10, 5000, 1000 and 25000 nM, respectively (Fig 4B; S2D and S2E Fig). In addition, 5000, 250, 250, 25000 and 1000 nM of luteolin, pyronaridine tetraphosphate, nimbolide, gedunin and enoxacin treatments significantly inhibited the growth of T. equi, respectively (Fig 4C and S3A Fig). Finally, the in vitro growth of B. caballi was significantly inhibited at 1000, 10, 250, 10000 and 1000 nM luteolin, pyronaridine tetraphosphate, nimbolide, gedunin and enoxacin treatments, respectively (Fig 4D and S3B Fig).

To determine the best effective drug against the in vitro growth of bovine and equine hemoparasites and examine whether IC50 values of compounds differ from each other, ranking of drug potencies were assessed. The IC50 of the diminazene aceturate determined by the fluorescence-based assay of each parasite was used as a control. The obtained IC50s from all treated groups showed different levels of statistical significant differences (P < 0.05) in comparison with diminazene aceturate treated group in vitro cultures of four parasites (Table 3). For B. bovis, luteolin and pyronaridine tetraphosphate treated groups showed the lowest statistical significant difference, followed by nimbolide, gedunin and enoxacin (Table 3). For B. bigemina, luteolin and nimbolide treated groups showed the lowest statistical difference in comparison with diminazene aceturate treated group followed by pyronaridine tetraphosphate, gedunin and enoxacin (Table 3). While, luteolin and gedunin showed the lowest statistical significant difference in comparison with diminazene aceturate treated group in vitro culture of T. equi, followed by pyronaridine tetraphosphate, nimbolide and enoxacin (Table 3). Finally, for B. caballi, pyronaridine tetraphosphate, nimbolide and enoxacin showed very statistical significant difference in comparison with diminazene aceturate treated group, while, other treated groups, exhibited an extremely significant difference (Table 3). These results suggest that luteolin has the best activity against the in vitro growth of bovine Babesia and T. equi parasites next to diminazene aceturate. While, for B. caballi pyronaridine tetraphosphate showed the best activity after diminazene aceturate. Other drugs including, nimbolide, gedunin and enoxacin were effective but with lower activity against the in vitro growth of bovine and equine hemoparasites.

Discussion

In recent years, antibabesial drugs commonly used in field, such as diminazene aceturate and imidocarb dipropionate [21], have shown some drawbacks, including toxicity and resistance [5]. Therefore, developing safer antibabesial medicaments merits priority in veterinary treatment research. However, several studies regarding the mode of action and target of new candidates that require more time, effort, and money should be applied. Therefore, the quest for future babesiosis drugs should be shifted to the in vitro screening of large-scale chemicals either commercially available or non-commercially available compounds. Then, the compounds that show potent antibabesial effects in vitro should be examined for their inhibitory effects in vivo before application in the field. In the current study, we attempted to establish an assay using SGI stain suitable for mass drug screening in vitro for B. bovis, B. bigemina, B. caballi, and T. equi.

The fluorescence-based assay has been approved for in vitro evaluation and drug screening for certain hemoprotozoan parasites [8]. The assay depends on DNA detection using SGI stain. This stain is an asymmetrical cyanine dye that has been used as a substitute for ethidium bromide in molecular biology for several years. Its mode of action depends on the interaction with double-stranded DNA, preferring G and C base pairs [22]. This interaction yields a highly fluorescent, absorbing light at a wavelength between 390 and 505 nm, with a peak at 497 nm and a secondary peak near 254 nm. It emits light at 505 to 615 nm, with a peak at 520 nm [http://www.sigmaaldrich.com/sigma-aldrich/datasheet/s9430dat.pdf]. The assay quality is determined by statistical parameters including the Z’-factor, S/N ratio, % CVmax, and % CVmin [19, 23, 24]. The Z’-factor represents the separation of the distributions between the positive and negative controls, and its value should be ≥ 0.5 [19]. Low values of the Z’ factor indicate that the assay format is not effective for generating useful data. However, higher values of the S/N ratio and % CVmin revealed the strength and accuracy of the assay in generating reliable data [19].

In the current study, we evaluated the usefulness of our modified fluorescence-based assay for mass drug screening for Babesia and Theileria parasites. The modifications, including the final volume of the medium, hematocrit, and the non-daily replacement of the medium, improved the performance of the assay and made it more practical for the large-scale screening of drugs. The results of the HTS assay for B. bovis with 2.5% HCT generated S/N ratios 1–10 and 12–203 times higher than those obtained from our previous laboratory assay [10] and those reported in malaria research [11], respectively. Meanwhile, the S/N ratios obtained from the assay with 5% HCT for equine Babesia and Theileria parasites were 6–16 times higher than those obtained in malaria research [11].

Furthermore, the IC50 values obtained from the fluorescence assay without daily replacement of medium at 2.5% and 5% HCTs for bovine Babesia and equine Babesia and Theileria parasites, respectively, for diminazene aceturate, luteolin, pyronaridine tetraphosphate, nimbolide, gedunin, and enoxacin were similar to those obtained by microscopy in our study and those obtained in previous reports [10, 20, 25]. Taken together, the consistency in IC50 values calculated by these two methods highlights the robustness of the current assay as an accurate, simple, and rapid method for mass drug screening in an in vitro culture.

The drugs luteolin, pyronaridine tetraphosphate, and enoxacin had not yet been tested against Babesia and Theileria species but have been previously documented to be effective against the growth of Plasmodium falciparum [26, 27, 28]. Nimbolide was previously used only against B. bovis [10], but its inhibitory effect against other Babesia and Theileria parasites had not yet been evaluated. The IC50 values of nimbolide for B. bovis and T. equi parasites were higher than the value calculated for P. falciparum 2000 nM (0.95 microgram/ml) [29]. In contrast, the IC50 values of nimbolide for B. bigemina and B. caballi parasites were, respectively, similar to and lower than that calculated for P. falciparum [29]. Luteolin is one of the flavonoids present in fruits, vegetables, wine, tea, and coffee [30]. In addition, this plant product has antioxidant, antitumor, anti-inflammatory, antimicrobial, antiviral, and antiprotozoal activities against Toxoplasma [31], Trypanosoma [32], Leishmania [33], and malaria [26]. The IC50 values of luteolin against the growth of 3D7 and 7G8 P. falciparum have been reported to be 11000 ± 1000 nM and 12000 ± 1000 nM, respectively [26]. These values appeared to be higher than those for bovine and equine Babesia and equine Theileria parasites determined by the current assay, which might indicate the susceptibility of Babesia parasites to this drug. Pyronaridine tetraphosphate is a Chinese drug that has a strong inhibitory effect against chloroquine-sensitive and-resistant strains of P. falciparum [34, 35]. The IC50 value of pyronaridine has been reported to be 2 nM against the growth of KT1 and KT3 P. falciparum [27], which is lower than the values obtained for Babesia and equine Theileria parasites in the current study. Enoxacin is one of the fluoroquinolones, a potent DNA gyrase inhibitor that had the lowest IC50 values against FCC1 and VNS strains of P. falciparum [28, 36]. The IC50 values of enoxacin for Babesia and Theileria parasites were significantly higher than the IC50 values for P. falciparum, which ranged from 2300 to 4400 nM [28, 37]. Finally, the obtained IC50 values of diminazene aceturate for B. caballi were lower than and nearly similar to those previously calculated by microscopy [38, 20], respectively.

In summary, our optimized fluorescence-based assay with 2.5% and 5% HCTs without daily replacement of the medium for bovine and equine Babesia and Theileria parasites, respectively, offers a new approach for accurate, simple, and rapid detection of Babesia and Theileria parasites and large-scale screening of antibabesial drugs in an in vitro culture. In addition, luteolin and pyronaridine tetraphosphate drugs exhibited the best growth inhibition of bovine and equine hemoparasites next to diminazene aceturate, fellowed by nimbolide, gedunin and enoxacin. These drugs might be more effective if used as part of a combination therapy rather than a single therapy. Further studies are required for analyzing the synergistic or an antagonistic effect of these drugs when used in combination with each other, and to determine the best effective composition ratio for the growth inhibition of bovine and equine hemoparasites for clinical application.

Supporting Information

S1 Fig. Fluorescence-based monitoring of luteolin, pyronaridine tetraphosphate, nimbolide, gedunin and enoxacin-induced growth inhibition of B. bovis during 4 days of treatment without daily replacement of medium.

(A) Fluorescence-based monitoring of luteolin-induced growth inhibition of B. bovis. (B) Fluorescence-based monitoring of pyronaridine tetraphosphate-induced growth inhibition of B. bovis. (C) Fluorescence-based monitoring of nimbolide-induced growth inhibition of B. bovis. (D) Fluorescence-based monitoring of gedunin-induced growth inhibition of B. bovis. (E) Fluorescence-based monitoring of enoxacin-induced growth inhibition of B. bovis. Statistically significant differences are indicated by asterisks (*P <0.05) between the drug-treated cultures and the control cultures. Each value represents the mean of triplicate wells after subtraction of the background fluorescence for non-parasitized RBCs.

https://doi.org/10.1371/journal.pone.0125276.s001

(PDF)

S2 Fig. Fluorescence-based monitoring of luteolin, pyronaridine tetraphosphate, nimbolide, gedunin and enoxacin-induced growth inhibition B. bigemina during 4 days of treatment without daily replacement of medium.

(A) Fluorescence-based monitoring of luteolin-induced growth inhibition of B. bigemina. (B) Fluorescence-based monitoring of pyronaridine tetraphosphate-induced growth inhibition of B. bigemina. (C) Fluorescence-based monitoring of nimbolide-induced growth inhibition of B. bigemina. (D) Fluorescence-based monitoring of gedunin-induced growth inhibition of B. bigemina. (E) Fluorescence-based monitoring of enoxacin-induced growth inhibition of B. bigemina. Statistically significant differences are indicated by asterisks (*P <0.05) between the drug-treated cultures and the control cultures. Each value represents the mean of triplicate wells after subtraction of the background fluorescence for non-parasitized RBCs.

https://doi.org/10.1371/journal.pone.0125276.s002

(PDF)

S3 Fig. Fluorescence-based monitoring of gedunin and enoxacin-induced growth inhibition of T. equi, and B. caballi on the fourth day of treatment.

(A) Correlation between gedunin and enoxacin concentrations and RFUs on T. equi. (B) Correlation between gedunin and enoxacin concentrations and RFUs on B. caballi. Each value represents a mean of triplicate wells after subtraction of the background fluorescence for non-parasitized RBCs. Statistically significant differences are indicated by asterisks (*P <0.05) between the drug-treated cultures and the control cultures.

https://doi.org/10.1371/journal.pone.0125276.s003

(PDF)

S4 Fig. The absolute values of growth inhibition for B. bovis by diminazene aceturate on the fourth day of treatment.

(A) Growth inhibition of B. bovis (x-axis) and log concentration of diminazene aceturate (nM) (y-axis) by the fluorescence-based method without daily replacement of the medium (black) and with daily replacement of the medium (gray). (B) Growth inhibition of B. bovis by diminazene aceturate on the fourth day estimated by the fluorescence-based method. (C) Growth inhibition of B. bovis by diminazene aceturate on the fourth day estimated by the microscope-based method. Each value is presented as the mean of three triplicate wells ± SD after subtraction of the background fluorescence for non-parasitized RBCs.

https://doi.org/10.1371/journal.pone.0125276.s004

(PDF)

S1 Table. Statistical parameters for determining the quality of the high-throughput screening (HTS) assay in B. bovis, B. bigemina, T. equi, and B. caballi parasites with different percentages of HCTs.

S/N ratio = Signal to noise, % CVmax = coefficient of variation at the maximum signal and % CVmin = coefficient of variation at the minimum signal.

https://doi.org/10.1371/journal.pone.0125276.s005

(DOCX)

Author Contributions

Conceived and designed the experiments: MAR SAESES MAY ESESES NY II. Performed the experiments: MAR SAESES MAT AE MO AS. Analyzed the data: MAR SAESES GE SEK MEA. Contributed reagents/materials/analysis tools: MAR SAESES NY II. Wrote the paper: MAR SAESES MAT SEK MAY ESESES GE NY II.

References

  1. 1. Uilenberg G. Babesia—a historical overview. Vet Parasitol. 2006; 31: 3–10.
  2. 2. Mehlhorn H, Schein E. Redescription of Babesia equi Laveran as Theileria equi. Parasitol Res. 1998; 84: 467–475. pmid:9660136
  3. 3. Schein E. Equine babesiosis. In: Rictic M. (Ed.), Babesiosis of Domestic Animals and Man. CRC Press, Boca Raton FL.; 1988. PP. 197–209.
  4. 4. Homer JM, Aguilar-Delfin I, Telford SR, Krause PJ, Persing DH. Babesiosis. Clin Microbiol Rev. 2000; 13: 451–469. pmid:10885987
  5. 5. Mosqueda J, Olvera-Ramirez A, Aguilar-Tipacamu G, Canto GJ. Current advances in detection and treatment of babesiosis. Curr Med Chem. 2012; 19: 1504–1518. pmid:22360483
  6. 6. Silva MG, Domingos A, Esteves MA, Cruz MEM, Suarez CE. Evaluation of the growth-inhibitory effect of trifluralin analogues on in vitro cultured Babesia bovis parasites. Int J Parasitol Drugs Drug Resist. 2013; 3: 59–68. pmid:24533294
  7. 7. Ruebush TK, Rubin RH, Wolpow ER, Cassaday PB, Schultz MG. Neurologic complications following the treatment of human Babesia microti infection with diminazene aceturate. Am J Trop Med Hyg. 1979; 28: 184–189. pmid:572145
  8. 8. Smilkstein M, Sriwilaijaroen N, Kelly JX, Wilairat P, Riscoe M. Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screening. Antimicrob Agents Chemother. 2004; 48: 1803–1806. pmid:15105138
  9. 9. Mayr LM, Fuerst P. The future of high-throughput screening. J Biomol Screen. 2008; 13: 443–448. pmid:18660458
  10. 10. Guswanto A, Sivakumar T, Rizk MA, Elsayed SA, Youssef MA, Elsaid EE, 1et al. Evaluation of a fluorescence-based method for antibabesial drug screening. Antimicrob Agents Chemother. 2014; 58 (8); 4713–4717. pmid:24914124
  11. 11. Hasenkamp S, Sidaway A, Devine O, Roye R, Horrocks P. Evaluation of bioluminescence-based assays of anti-malarial drug activity. Malar J. 2013; 12: 58. pmid:23394077
  12. 12. Hines SA, Palmer GH, Jasmer DP, McGuire TC, McElwain TF. Neutralization-sensitive merozoite surface antigens of Babesia bovis encoded by members of a polymorphic gene family. Mol Biochem Parasitol. 1992; 55: 85–94. pmid:1279421
  13. 13. Hotzel I, Suarez CE, McElwain TF, Palmer GH. Genetic variation in the dimorphic regions of RAP-1 genes and rap-1 loci of Babesia bigemina. Mol Biochem Parasitol. 1997; 90: 479–489. pmid:9476795
  14. 14. Avarzed A, Igarashi I, Kanemaru T, Hirumi K, Omata Y, Saito A. Improved in vitro cultivation of Babesia caballi. J Vet Med Sci. 1997; 59: 479–481. pmid:9234227
  15. 15. Bork S, Yokoyama N, Ikehara Y, Kumar S, Sugimoto C, Igarashi I. Growth-inhibitory effect of heparin on Babesia parasites. Antimicrob Agents Chemother. 2004; 48: 236–241. pmid:14693545
  16. 16. Igarashi I, Njonge FK, Kaneko Y, Nakamura Y. Babesia bigemina: In vitro and in vivo effects of curdlan sulfate on growth of parasites. Exp Parasitol. 1998; 90: 290–293. pmid:9806875
  17. 17. AbouLaila M, Nakamura K, Govind Y, Yokoyama N, Igarashi I. Evaluation of the in vitro growth inhibitory effect of epoxomicin on Babesia parasites. Vet Parasitol. 2010; 167: 19–27. pmid:19896277
  18. 18. Johnson JD, Dennull RA, Gerena L, Lopez-Sanchez M, Roncal NE, Waters NC. Assessment and continued validation of the malaria SYBR Green I-based fluorescence assay for use in malaria drug screening. Antimicrob Agents Chemother. 2007; 51: 1926–1933. pmid:17371812
  19. 19. Zhang JH, Chung TD, Oldenburg KR. A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J Biomol Screen. 1999; 4: 67–73. pmid:10838414
  20. 20. AbouLaila M, Munkhjargal T, Sivakumar T, Ueno A, Nakano Y, Yokoyama M, et al. Apicoplast-targeting antibacterials inhibit the growth of Babesia parasites. Antimicrob Agents Chemother. 2012; 56: 3196–3206. pmid:22391527
  21. 21. Kuttler KL. In Babesiosis. Ristic M; Keire J.P. (Eds.), Academic Press: New York; 1981; 25–63.
  22. 22. Bennett TN, Paguio M, Gligorijevic B, Seudieu C, Kosar AD, Davidson E, et al. Novel, rapid, and inexpensive cell-based quantification of antimalarial drug efficacy. Antimicrob Agents Chemother. 2004; 48: 1807–1810. pmid:15105139
  23. 23. Makler M, Ries L, Ries J, Horton R, Hinrichs D. Detection of Plasmodium falciparum infection with the fluorescent dye, benzothiocarboxypurine. Am J Trop Med Hyg. 1991; 44: 11–16. pmid:1996734
  24. 24. Skeidsvall J, Veland P. Analysis of double-stranded DNA capillary electrophoresis laser-induced fluorescence detection using the monomeric dye SYBR Green I. Anal Biochem. 1995; 231: 359–365. pmid:8594986
  25. 25. Azirwan G, Thillaiampalam S, Muncharee T, Yokoyama N, Igarashi I. In vitro inhibitory effect of gedunin on Babesia and Theileria parasites. J Protozool Res. 2013; 23: 1–6.
  26. 26. Lehane AM, Saliba KJ. Common dietary flavonoids inhibit the growth of the intraerythrocytic malaria parasite. BMC Research Notes. 2008; 1: 26. pmid:18710482
  27. 27. Chavalitshewinkoon-Petmitr P, Pongvilairat G, Auparakkitanon S, Wilairat P. Gametocytocidal activity of pyronaridine and DNA topoisomerase II inhibitors against multidrug-resistant Plasmodium falciparum in vitro. Parasitol Int. 2000; 48: 275–280. pmid:10725690
  28. 28. Divo AA, Sartorelli AC, Patton CL, Bia FJ. Activity of fluoroquinolone antibiotics against Plasmodium falciparum in vitro. Antimicrob Agents Chemother. 2008; 8: 1182–1186.
  29. 29. Rochanakij S, Thebtaranonth Y, Yenjai C, Yuthavong Y. Nimbolide, a constituent of Azadirachta indica, inhibits Plasmodium falciparum in culture. Southeast Asian J Trop Med Public Health. 1985; 16: 66–72. pmid:3895455
  30. 30. Havsteen BH. The biochemistry and medical significance of the flavonoids. Pharmacol Ther. 2002; 96 (2–3): 67–202. pmid:12453566
  31. 31. Weiss LM, Ma YF, Takvorian PM, Tanowitz HB, Wittner M. Bradyzoite development in Toxoplasma gondii and the hsp70 stress response. Infect Immun. 1998; 66: 3295–3302. pmid:9632598
  32. 32. Mamani-Matsuda M, Rambert J, Malvy D, Lejoly-Boisseau H, Daulouede S, Thiolat D, et al. Quercetin induces apoptosis of Trypanosoma brucei gambiense and decreases the proinflammatory response of human macrophages. Antimicrob Agents Chemother. 2004; 48: 924–929. pmid:14982785
  33. 33. Sen G, Mandal S, Saha Roy S, Mukhopadhyay S, Biswas T. Therapeutic use of quercetin in the control of infection and anemia associated with visceral leishmaniasis. Free Radic Biol Med. 2005; 38: 1257–1264. pmid:15808423
  34. 34. Zheng XY, Chen C, Gao FH, Zhu PE, Guo HZ. Synthesis of new antimalarial drug pyronaridine and its analogues. Acta Pharm Sin. 1982; 17: 118–125.
  35. 35. Fu S, Bjorkman A, Wahlin B, Ofori-Adjei D, Ericsson O, Sjoqvist F. In vitro activity of chloroquine, the two enantiomers of chloroquine, desethylchloroquine and pyronaridine against Plasmodium falciparum. Br J Clin Pharmacol. 1986; 22: 92–96.
  36. 36. Overington JP, Al-Lazikani B, Hopkins AL. How many drug targets are there? Nat Rev Drug Discov. 2006; 5: 993–996. pmid:17139284
  37. 37. Nassira M, Liliane C, Jean-François F, Khemais F, Olivier S, Eling W, et al. In vitro activities of 25 quinolones and fluoroquinolones against liver and blood stage Plasmodium spp. Antimicrob Agents Chemother. 2003; 47: 2636–2639. pmid:12878530
  38. 38. Salama A, AbouLaila M, Moussa A, Nayelb A, El-Sify A, Terkawi MA, et al. Evaluation of in vitro and in vivo inhibitory effects of fusidic acid on Babesia and Theileria parasites. Vet Parasitol. 2013; 191: 1–10. pmid:22985928