Quercetin inhibits Toxoplasma gondii tachyzoite proliferation and acts synergically with azithromycin
HomeHome > Blog > Quercetin inhibits Toxoplasma gondii tachyzoite proliferation and acts synergically with azithromycin

Quercetin inhibits Toxoplasma gondii tachyzoite proliferation and acts synergically with azithromycin

Jun 05, 2024

Parasites & Vectors volume 16, Article number: 261 (2023) Cite this article

554 Accesses

Metrics details

Quercetin (QUE) is a natural polyphenol known to have numerous pharmacological properties against infectious and non-infectious diseases. Azithromycin (AZ) is an antibiotic that belongs to the azalide class of antimicrobials and an antiparasitic that is known to be effective in combination with clindamycin against pyrimethamine/sulfadiazine-resistant Toxoplasma gondii tachyzoites in clinical settings. Both compounds are known to target protein synthesis and have anti-inflammatory properties. However, little is known about QUE and AZ synergistic interaction against T. gondii growth. Here, we report for the first time the effects of the combination of QUE and AZ on T. gondii growth. The 50% inhibitory concentration (IC50) for QUE at 72 h of interaction was determined to be 0.50 µM, whereas AZ gave an IC50 value of 0.66 µM at 72 h of interaction with parasites. Combination testing of QUE and AZ in a ratio of 2:1 (QUE:AZ) showed an IC50 value of 0.081 µM. Interestingly, a fractional inhibitory index value of 0.28 was observed, indicating a strong synergy. QUE was also found to upregulate the generation of reactive oxygen species and cause dysfunction of the mitochondria membrane of both intracellular and extracellular T. gondii tachyzoites. Overall, the results indicate that QUE is a novel lead capable of synergizing with AZ for inhibiting T. gondii growth and may merit future investigation in vivo for possible combination drug development.

Toxoplasmosis is a global neglected parasitic disease caused by the obligatory intracellular protozoan parasite Toxoplasma gondii. The disease is known to affect both warm- and cold-blooded host cells [1,2,3] and it has been reported that more than one-third of the world population is infected with T. gondii [1, 3], including about 40 million people in the USA [4]. Also of major concern is that there are about 1.20 million cases of congenital toxoplasmosis across the world [5], with about 400 to 4000 cases reported annually in the USA [4]. The number may be higher since not all US states perform T. gondii seroprevalence testing in pregnant women. In fact, only a few states, such as Massachusetts and New Hampshire, screen for congenital toxoplasmosis during prenatal visits [6, 7].

Toxoplasma gondii infection in most immunocompetent individuals is clinically asymptomatic. However, immunocompromised people (e.g. HIV-infected patients, cancer patients, patients having organ transplants and those receiving blood transfusion) do present clinical signs that can range from mild to life threatening [4, 8, 9].

Toxoplasma gondii is acquired through the ingestion of tissue cysts in raw or uncooked meat, contaminated soils, water and contaminated food products [2]. The two first-line treatment for T. gondii infection in humans are combination therapy with sulfadiazine (SDZ) and pyrimethamine (PYR) and combination therapy with trimethoprim and sulfamethoxazole [10, 11]. However, these drugs are associated with serious adverse health effects, such as induction of leucopenia, neutropenia, hypersensitivity reactions, thrombocytopenia, bone marrow suppression and megaloblastic anemia, in patients, with severely low platelet count [10,11,12,13,14,15,16,17,18]. These challenges necessitate more research into finding new anti-T. gondii inhibitors against toxoplasmosis that can work as sole therapy or in combination with other drugs.

Azithromycin (AZ) is a macrolide antibiotic that is a derivative of erythromycin and which has been extensively researched and proven to be an effective anti-Toxoplasma agent in the event of treatment failure with the pyrimethamine/sulfadiazine combination. This antibiotic is structurally similar to erythromycin, with high antibacterial properties and a desirable pharmacokinetic profile. AZ has been demonstrated to block translation in the normally translationally active plasmodial apicoplast and thus has been generally applied in treating pneumonia and chlamydia, especially in pregnant women [18]. Furthermore, AZ has been shown to control T. gondii infection in human villous explants [19].

Quercetin (QUE) is a polyphenolic flavonoid found in most plant foods, herbal supplements, vegetables, fruits, tea, red wines and seeds. It contains a broad spectrum of biological properties that have protective abilities, such as anti-inflammatory [20, 21], anti-mutagenicity [22], anti-cancer [23] and anti-oxidization [24] effects, inherent antibacterial properties against Escherichia coli [25] and reducing or preventing cardiovascular diseases [26]. Previous studies have shown that QUE synergizes with amoxicillin in the killing of Staphylococcus epidermidis [27], as well as with other antibiotics, such as with levofloxacin, ceftriaxone, gentamycin, tobramycin and amikacin against Pseudomonas aeruginosa [28] and with epigallocatechin gallate against Leishmania [29]. However, little is known about the interaction of QUE with AZ against T. gondii growth.

In the present study, we tested QUE alone and its combination with AZ, to test the hypothesis of whether synergy would result.

Vero cells (a lineage originally isolated from kidney epithelial cells extracted from an African green monkey) expressing firefly luciferase (Luc2p) were obtained from NIH Biodefense and Emerging Infections Research Resources Repository (BEI Resources, Manassas, VA, USA; NAID; NIH; NR-10385). Toxoplasma gondii type I virulent strain RH-(YFP)2, expressing yellow fluorescent protein (YFP)2, was kindly provided by Prof. William H. Witola (College of Veterinary Medicine, University of Illinois, Urbana Champaign, IL, USA). Vero cells were grown in a T-25-cm3 flask containing Dulbecco’s Modified Eagle Medium (DMEM) (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 1% penicillin–streptomycin (PS) and amphotericin B solution as antibiotics (Gibco, Thermo Fisher Scientific) and 10% fetal bovine serum (FBS) (Life Technologies Inc., Thermo Fisher Scientific. Cultures were maintained at 37ºC with 5% CO2 to become confluent. Toxoplasma gondii tachyzoites of virulent strains RH-Wild-type (WT) with no fluorescent tag and hTERT (fibroblast) cells were provided by Prof. Silvia NJ Moreno (University of Georgia, Athens, GA, USA).

Vero cells (6 × 104 cells/200 µl) were seeded into black 96 well plates and incubated for 24 h at 37 °C with 5% CO2. At 24 h, the Vero cells were washed to remove dead cells and 100 µl of growth media added to the cells. QUE and AZ were added in a 100-µl volume at concentrations of 25, 12.5, 6.25, 3.12, 1.56 and 0 µM, respectively, and incubated for 48 h. After 48 h, 10 µl Alamar blue dye (Abcam, Waltham, MA, USA) was added to the culture wells, and the plates were covered with aluminum foil and incubated at standard culture conditions. The fluorescence was measured at 560/630 nm using the Tecan 200F infinite fluorescent plate reader (Tecan Group, Männedorf Switzerland). Experiments were performed in three independent experiments and the results presented as the mean ± standard deviation (SD) (n = 3).

The QUE and AZ were obtained from Santa Cruz Biotechnology Inc. (Dallas, TX, USA and prepared in dimethyl sulfoxide (DMSO). The T. gondii RH-(YFP)2 tachyzoites expressing yellow fluorescent protein (YFP)2 throughout culture were used to test the inhibitory effect of QUE alone and in combination with AZ on T. gondii parasites in vitro. Vero cells (6.0 × 104 cells/(200 µl) were seeded into 96-well plates and incubated at 37 °C with 5% CO2 for 24 h for 90% confluence, following which precisely 100 µl of freshly purified tachyzoites at a concentration of 1 × 104 parasites/well was added to the Vero cells. The experimental compound QUE and the standard drug (AZ) were added in a volume of 100 µl at concentrations 0, 1.56, 3.12, 6.25, 12.5 and 25 µM, respectively, and incubated at 37 °C with 5% CO2. Toxoplasma gondii growth at 72 of culture was measured using Tecan 200 F infinite fluorescent plate reader with excitation set at 485 nm and emission set at 535 nm (Tecan Group). The fluorescent intensities were converted into percentage inhibition using the formula reported in [30]. The concentrations of the compound QUE and AZ were plotted against the percentage inhibition of T. gondii growth using GraphPad Prism software 9.2 (GraphPad Software, San Diego, CA, USA. Experiments were conducted in three independent experiments and the results presented as the mean ± SD (n = 3).

At 72 h post-treatment with QUE and AZ, the plates were washed 3 times with 1× phosphate-buffered saline (PBS) to remove all compounds and all extracellular parasites. After washing, 100 µl of 10% FBS-supplemented DMEM was added to each well, followed by initial reading as day 0. Parasite fluorescence intensity was recorded for the following 72 h, and the 50% effective minimum concentrations were determined at 72 h to determine whether the compounds removed still had any effect on tachyzoites growth. The negative control wells (where no drugs were previously added) served as the 100% benchmark and the post-treatment with QUE and AZ withdrawal served as the experimental wells. The IC50s values were determined as stated above in the individual growth inhibition assay. Experiments were conducted in three independent experiments and the results presented as the mean ± SD of triplicate trials (n = 3).

Vero cells (6.0 × 104 cells/200 µl) were seeded into 96-well plates for 24 h, as described above for the testing of individual compounds, followed by purification of RH-(YFP)2 tachyzoites as described above. The concentration of parasites used in the individual studies described above was added at a volume of 100 µl, followed by the addition of drugs in a ratio of 1:1 (50 µM QUE: 50 µM AZ), 2:1 (50 µM QUE: 25 µM AZ) and 1:2 (25 µM QUE: 50 µM AZ), serially diluted, and parasite growth was monitored using the Tecan 200F infinite fluorescent plate reader (Tecan Group) with the filters set to 458 nm/535 nm as excitation and emission wavelengths, respectively. Parasite growth was monitored for 72 h, and the IC50s values were calculated from the interaction data at 72 h using GraphPad prism software version 9.2.0 (GraphPad Software). Experiments were conducted in three independent experiments and the results presented as the mean ± SD of triplicate trials (n = 3). To decipher the combination that could exert synergy and addictiveness for future modification, we used the fractional inhibitory concentration (FICI). Synergy was defined as an inhibition IC50 produced by a combination of compounds (QUE-AZ) that is greater than the sum of the inhibitory concentration effects produced by QUE or AZ alone. FICI values were determined the using formula reported in [31,32,33] : FICI = IC50p of QUE in combination (QUE-AZ [1:1, 1:2 or 2:1]/IC50p of QUE alone + IC50p of AZ in combination (QUE-AZ [1:1, 1:2 and 2:1])/IC50p of AZ alone. We used a standard guideline which states that FICI values < 0.5 are considered to be synergistic; FICI values ≥ 1 indicate an additive and FICI values ≥ 2 indicate an antagonistic interaction [31].

QUE has been previously reported to induce reactive oxygen species (ROS) production in Leishmania amazonensis. To verify whether QUE had any effect on ROS production in T. gondii tachyzoites, we used a procedure similar to that reported in an earlier study [30] with modifications. WT T. gondii tachyzoites provided by Silvia NJ Moreno (University of Georgia, Athens, GA, USA) were maintained in growth media without phenol red in intact cells. We harvested freshly lysed RH-Wild type (RH-W) parasites by passing them through a 27-gauge needle followed by filtration through a 3-µm filter. RH-W parasites (1.60 × 106 parasites/50 µl per well) were seeded into black 96-well plates and treated either with H2O2 (500 µM) as a positive control [34] or with QUE at different concentrations (0.66 and 12.5 µM), for 30 min at 37 °C with 5% CO2. After a 30-min incubation, ROS dye (Abcam) was added and the wells incubated for a further 45 min according to the manufacturer’s protocol and previous work [35]. Fluorescence intensities of the wells were measured at an excitation of 485 nm and emission at 563 nm, respectively, using the Tecan 200F infinite microplate reader (Tecan Group). Experiments were conducted in three independent experiments and the results presented as the mean ± SD of triplicate trials (n = 3).

For mitochondrial membrane potential (ΔΨm) measurements, we used the cationic JC-1 dye as a fluorescent probe (Abcam), as previously described [30, 36]. This membrane potential kit has been widely used in T. gondii tachyzoite mitochondrial membrane potential testing in vitro [35, 37,38,39,40], with modifications. We measured both the intracellular and extracellular ΔΨm of the parasite. For the intracellular assay, 6 × 104 Vero cells were seeded into wells of a black 96-well plate with a clear bottom (Costar, Corning Inc., Corning, NY, USA) until they were confluent. Then 6 × 104 tachyzoites from T. gondii RH-WT strains were added. In contrast, in the assay for extracellular parasites, 1 × 105 freshly purified parasites in 100 μl media were seeded directly into the wells (no host cells). Then, either 100 μl solution containing 0.625, and 16.67 μM of QUE as the experimental drug, 50 μM of carbonyl cyanide m-chlorophenyl hydrazone (CCCP; Alfa Aesar, Haverhill, MA, USA) as a positive control or 1× HBSS (assay buffer) the negative control was added to the designated wells and incubated for 8 h at 37 °C with 5% CO2 [30, 36]. A 10-μl aliquot of JC-1 was added to the wells, and the plates were covered with aluminum foil and incubated for 45 min. The solutions were then removed and centrifuged at 12 °C, 2000 rpm for 5 min, and the supernatant was removed. Next, 100 μl of assay buffer was added to each well and centrifuged again under the same conditions and the supernatant discarded. The parasite pellets were resuspended in solution with 100 μl of assay buffer for the extracellular assay. For the intracellular assay, the supernatant was discarded followed by the addition of 100 μl of assay buffer. Both intracellular and extracellular parasite assays were imaged using an EVOS FL fluorescence microscope (Invitrogen Life Technologies, Thermo Fisher Scientific). The experiments were performed in triplicate (n = 3) [36].

To further validate the results of our mitochondria assay, we used a modified protocol [30, 37, 39]. Fresh extracellular RH-RFP parasites (2.84 × 104) were incubated in a complete medium containing QUE at concentrations of 0.62 and 16.67 µM, in complete medium without the drug (medium only with parasites as negative control) or in medium containing 500 µM H2O2 as a negative control. After 8 h of incubation under the standard culture conditions of 5% CO2 at 37 °C, the ATP Detection Assay Kit - Luminescence (catalog no. 700410; Cayman Chemical, Ann Arbor, MI, USA) was used to measure the luminescence of samples. QUE (the drug treated), the positive control (H2O2) and the negative control (medium with parasites without drugs) were washed twice with PBS and the pellets lysed on ice with 100 µl of lysis buffer. Next, 10 µl of the parasite’s lysates were added to 100 µl of adenosine triphosphate (ATP) detection working solution in each opaque microplate’s well (Corning). The plates were incubated for 20 min at room temperature according to the protocol of the ATP Detection Assay Kit, the covers of the plates were removed and luminescence was measured using the BioTek Cytation cell imaging multi-mode microplate reader with software Gen 5.3.1 (Agilent Technologies Inc., Santa Clara, CA, USA). The effect of treatments on T. gondii ATP luminescence was compared with ATP luminescence of untreated T. gondii population (medium with parasites without experimental drugs or negative controls) and expressed as percentages of these control ATP values. Experiments were conducted in three independent experiments and the results presented as the mean ± SD of triplicate trials.

GraphPad Prism software (GraphPad Software) was used to determine the IC50 values. A one-way analysis of variance (ANOVA) analysis was used to distinguish any statistical differences. A p value of 0.05 was considered to indicate significance.

The IC50 values for QUE and AZ against T. gondii tachyzoite growth at 72 h were determined to be 0.50 µM and 0.66 µM, respectively (Table 1).

The growth curves of the parasite, as percentage growth inhibition, in the presence of different concentrations of AZ and QUE, respectively, are presented in Fig. 1a, b. Interestingly, the IC50s values in our current study for both individual compounds are comparatively similar to those typical of the currently used drugs PYR and SDZ reported using HFF cells (0.95–1.55 µM for SDZ and 2.42–3.52 µM for PRY [30]; 1.17 ± 0.076 for PRY [41]); 0.79–1.5 µM for PYR [42]; 3.4 µM for PYR [32]; 0.8 µM for PYR [43]; 0.16 µM for PYR [44]).

Toxoplasma gondii growth inhibition curves at 72 h in the presence of quercetin (a) and azithromycin (b) at concentrations of 0, 1.56, 3.12, 6.25, 12.5 and 25 µM, interaction. Data are presented as means (filled circles) of three independent experiments performed in triplicate, with the standard error of the mean (whiskers)

Combined treatment with QUE and AZ was performed to determine whether these drugs could synergize against T. gondii growth in vitro. We noted that the combined treatment with QUE and ZA at a ratio of 2:1 (QUE: AZ) showed a strong synergy at 72 h after treatment initiation. The IC50s values for combined treatment with QUE and ZA at a ratio of 2:1 (QUE: AZ), 1:2 (QUE: AZ) and 1:1 of (QUE: AZ) were determined to be 0. 89, 1.50 and 0.081 µM, respectively (Table 1). The fractional inhibitory concentrations were calculated to be 0.28. The ratio of 2:1 (QUE: AZ) was strongly synergistic based on the FICI calculated (Table 1). However, combined treatment with QUE and AZ at ratios of 1:2 (QUE: AZ) and 1:1 (QUE: AZ) resulted in antagonistic interactions. The growth curve for the 2:1 ratio of QUE: AZ is shown in Fig. 2.

In vitro T. gondii strain RH-(YFP)2 inhibitory growth curve at 72 h in the presence of combined quercetin (QUE) and azithromycin (AZ) at the ratio of 2:1 (QUE: AZ) (50 µM QUE: 25 µM AZ). Concentrations tested were 50, 25, 12.5, 6.25, 3.125 and 0 µM for QUE, and 25, 12.5, 6.25, 3.125, 1.56, and µM for AZ. Data are presented as the means (filled circles) of three independent experiments performed in triplicate, with the standard error of the mean (whiskers). Q+A (2.1), Quercetin: azithromycin at the ratio of 2:1

The 50% cytotoxic concentrations (CC50) were calculated to be 14.98 µM for QUE, 2575 µM for AZ, 60.82 µM for QUE+AZ(2:1), 47.17 µM for QUE+AZ (1:2) and 37.22 µM for QUE+AZ (1:1) (Table 1). The selectivity indices (SI) for QUE, AZ and QUE+AZ (2:1) were calculated and are presented in Table 1. The SI values for the 1:1, 1:2 and 2:1 ratios of the QUE+AZ combinations were calculated to be 31, 42, and 760.25, respectively. All of the individual compounds and ratios tested had a broad-spectrum inhibitory effect on parasite growth in vitro compared to some of the current drugs used in the clinical setting to treat T. gondii infection.

To decipher whether the withdrawal of QUE after 72 h of treatment had any effect on the continued growth of tachyzoites during the first 72 h post-drug withdrawal, we determined the curves for parasite inhibition (Fig. 3a, b). It was noted that withdrawal of the compound did not abolish its inhibitory effect on parasite growth relative to the standard drug (AZ) tested at 72 h (Table 2). The IC50s values were calculated to be 0.49 and 0.20 µM for QUE and AZ at 72 h, respectively (Table 2).

Toxoplasma gondii inhibition curves for parasites first treated for 72 h and then grown in growth media for 72 h after drug withdrawal. a and b Depict growth curve of Quercetin and Azithromycin withdrawal treatment. Data are presented as the mean (filled circles) of three independent experiments performed in triplicate, with the standard error of the mean (whiskers)

AZ has also been known to have an effect on a secondary target of parasites through the apicoplast [45, 46]. Specifically, it has been reported that AZ affects lipid levels and membrane lipid fluidity in cells [47, 48]. Taken together, our findings and previous data suggest that AZ could affect sphingolipid and phospholipid production in the apicoplast, which is crucial for the parasite lytic cycle.

QUE causes high ROS production and mitochondrial membrane disruption, which may affect lipid synthesis (e.g. phospholipids and sphingolipids) directly or indirectly in the apicoplast and the mitochondria. Lipids are crucial for T. gondii invasion, proliferation and modulation of intracellular calcium that triggers parasites egress and controls most of the lytic cycle activities in the T. gondii parasite. However, this conjecture requires further study.

Our observation of the effect of AZ remaining active against tachyzoite growth even after drug withdrawal confirmed its long-acting ability due to its high half-life > 50 h in host cells [49].

The mitochondria is considered to be the powerhouse of eukaryotic cells, performing various functions, including ROS mediation, ATP production, fatty acid synthesis, translation and transcription [50]. To better understand the mechanism of action of QUE on ΔΨm, ATP production and ROS production, we employed assays that measured intracellular parasite responses to QUE in a dose-dependent manner. The results showed the effect of QUE on ΔΨm (see Fig. 4a). Similarly, we observed that QUE disrupted extracellular parasite ΔΨm (Fig. 4b). Also, to explore the effect of QUE on parasite ATP production in vitro, we performed an ATP production assay (see results of assay in Fig. 5). Study of the generation of ROS was also carried out to determine the effect of QUE on ROS released (see results of assay in Fig. 6). We observed slight statistical differences between 25 µM of QUE versus 50 µM of H2O2 (p < 0.026) and 12.5 µM of QUE versus 50 µM of H2O2 (p < 0.026), confirming results reported in a previous study in Leishmania amazonensis which showed that QUE causes ROS production and mitochondrial dysfunction [50]. Also, several studies have shown that compounds that lead to ΔΨm disruption and high ROS production could lead to disruption of ATP production in parasites [30, 37, 39, 51]. Furthermore, compounds exerting high ROS production and mitochondria membrane depolarization lead to peroxidation of long-chain fatty acids and the production of toxic intermediates, such hexanal, aldehyde and alkenes in host cells and parasite cells, further resulting in apoptotic activities and eventually cell death [52, 53]. We observed a statistical difference between the treatments with QUE, H2O2 (positive control) and medium alone (negative control). Confirming the results of previous studies reported in Lesihmania spp, using QUE, our current study showed a statistical difference between the standard positive control (50 µM H2O2) versus 0.62 µM QUE (p < 0.001), 16.67 µM QUE versus 0.62 µM QUE (p < 0.001), medium versus 50 µM H2O2 and 16.67 µM QUE versus 0.62 µM QUE (p < 0.001). Interestingly, the IC50 of QUE alone and in combinations with AZ were observed to be effective at submicrolar concentrations, which is in contrast to the results reported for Leishmania spp. [50]. This difference could be attributed to the host cell types used as a medium of propagation of the parasites, the concentration of QUE and/or the number of parasites used. It has also been reported that QUE is highly anti-inflammatory and antioxidant, and these properties have been associated with its oxidative, kinase and cell-cycle inhibition [54]. Studies have also shown its ability to induce apoptosis in cancer cells [55]. Our previous work using dihydroquercetin showed T. gondii inhibition but not as effective as QUE [32], possibly partly due to the number of hydroxyl groups available in QUE and the analog dihydroquercetin.

a Quercetin disrupts the mitochondria membrane potential (MMP) in extracellular T. gondii tachyzoites. White arrows indicate parasites. The panel with red represents the uncompromised MMP in parasites and the panel with green indicates the compromised MMP in parasites treated with Que (0.62 and 16.67 µM) and a standard compound (CCCP) at 50 µM. The green indicates compromised MMP. Scale bar: 200 µm. b Que disrupts the MMP in intracellular T. gondii tachyzoites. Small white arrows indicate parasites, and short large arrowheads indicate host cells. The red spikes indicate intact mitochondria in tachyzoites treated with Que (0.62 and 16.67 µM) and a standard compound CCCP at 50 µM. The green indicates the compromised MMP. Scale bar: 200 µm. CCCP, Carbonyl cyanide m-chlorophenyl hydrazone; Que, quercetin

Quercetin disrupts ATP production in extracellular T. gondii tachyzoites. QUE, Quercetin. ** and *** indicates a statistical difference between treatments at p < 0.01 and p < 0.001 respectively

Quercetin induces ROS production in intracellular T. gondii tachyzoites. H2O2, Hydrogen peroxide; QUE, quercetin; ROS, reactive oxygen species. * indicates p < 0.05

In conclusion, we found that QUE inhibited tachyzoite growth and also caused disruption of the mitochondrial membrane potential, depletion of ATP and increased ROS production. Any organelle disruption could directly or indirectly affect energetics and many other pathways, depending on the mitochondria-directed pathways. Comparatively, the combination therapy (AZ + QUE) was more effective against parasite proliferation at a ratio of 2:1 (QUE: AZ) than each of the individual compounds alone. This implies that QUE might be a good candidate for future combination with AZ. In vivo testing to ascertain its efficacy and safety will be an exciting and necessary investigation to undertake.

All data has been included in the manuscript and raw data are available upon request.

Azithromycin

Half-maximal inhibitory concentration

Quercetin

Pappas G, Roussos N, Falagas ME. Toxoplasmosis snapshots: global status of Toxoplasma gondii seroprevalence and implications for pregnancy and congenital toxoplasmosis. Int J Parasitol. 2009;39:1385–94.

Article PubMed Google Scholar

Dubey JP. Toxoplasmosis of animals and humans. Boca Raton: CRC Press; 2022.

Google Scholar

Flegr J, Prandota J, Sovičková M, Israili ZH. Toxoplasmosis—a global threat correlation of latent toxoplasmosis with specific disease burden in a set of 88 countries. PLoS ONE. 2014;9:90203.

Article Google Scholar

Centers for disease control prevention. Parasites-toxoplasmosis (Toxoplasma Infection): epidemiology and risk. 2022. https://www.cdc.gov/parasites/toxoplasmosis/epi.html. Accessed 19 Aug 2022.

Rostami A, Karanis P, Fallahi S. Advances in serological, imaging techniques and molecular diagnosis of Toxoplasma gondii infection. Infection. 2018;46:303–15.

Article CAS PubMed Google Scholar

Guerina NG, Hsu HW, Meissner HC, Maguire JH, Lynfield R, Stechenberg B, et al. Neonatal serologic screening and early treatment for congenital Toxoplasma gondii infection. The New England Regional Toxoplasma working group. N Engl J Med. 1994;330:1858–63.

Article CAS PubMed Google Scholar

Kota AS, Shabbir N. 2021. Congenital toxoplasmosis. Accessed 24 July 2022.

Fallahi S, Rostami A, Shiadeh MN, Behniafar H, Paktinat S. An updated literature review on maternal-fetal and reproductive disorders of Toxoplasma gondii infection. J Gynecol Obstet Hum Reprod. 2018;47:133–40.

Article CAS PubMed Google Scholar

Weiss LM, Kim K. Toxoplasma gondii: the model apicomplexan. Perspectives and methods. New York: Elsevier; 2022.

Google Scholar

Montoya JG, Boothroyd JC, Kovacs JA. Toxoplasma gondii. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s principles and practice of infectious diseases. 8th ed. Philadelphia: Churchill Livingstone Elsevier; 2017.

Google Scholar

Montazeri M, Mehrzadi S, Sharif M, Sarvi S, Tanzifi A, Aghayan SA, Daryani A. Drug resistance in Toxoplasma gondii. Front Microbiol. 2018;9:2587.

Article PubMed PubMed Central Google Scholar

Porter SB, Sande MA. Toxoplasmosis of the central nervous system in the acquired immunodeficiency syndrome. N Engl J Med. 1992;327:1643–8.

Article CAS PubMed Google Scholar

Rajapakse S, Chrishan Shivanthan M, Samaranayake N, Rodrigo C, Deepika FS. Antibiotics for human toxoplasmosis: a systematic review of randomized trials. Pathog Glob Health. 2013;107:162–9.

Article CAS PubMed PubMed Central Google Scholar

Carlier Y, Truyens C, Deloron P, Peyron F. Congenital parasitic infections: a review. Acta Trop. 2012;121:55–70.

Article PubMed Google Scholar

Ben-Harari RR, Goodwin E, Casoy J. Adverse event profile of pyrimethamine-based therapy in toxoplasmosis: a systematic review. Drugs R D. 2017;17:523–44.

Article CAS PubMed PubMed Central Google Scholar

Shammaa AM, Powell TG, Benmerzouga I. Adverse outcomes associated with the treatment of Toxoplasma infections. Sci Rep. 2021;11:1–8.

Article Google Scholar

Secrieru A, Costa IC, O’Neill PM, Cristiano ML. Antimalarial agents as therapeutic tools against toxoplasmosis—a short bridge between two distant illnesses. Molecules. 2020;25:1574.

Article CAS PubMed PubMed Central Google Scholar

Shiojiri D, Kinai E, Teruya K, Kikuchi Y, Oka S. Combination of clindamycin and azithromycin as alternative treatment for Toxoplasma gondii Encephalitis. Emerg Infect Dis. 2019;25:84.

Article Google Scholar

Castro-Filice LS, Barbosa BF, Angeloni MB, Silva NM, Gomes AO, Alves CMOS, Silva DAO, et al. Azithromycin is able to control Toxoplasma gondii infection in human villous explants. J Trans Med. 2014;12:1–12.

Article Google Scholar

Lotito SB, Zhang W, Yang CS, Crozier A, Frei B. Metabolic conversion of dietary flavonoids alters their anti-inflammatory and antioxidant properties. Free Radical Biol Med. 2011;51:454–63.

Article CAS Google Scholar

Azeem M, Hanif M, Mahmood K, Ameer N, Chughtai FRS, Abid U. An insight into anticancer, antioxidant, antimicrobial, antidiabetic and anti-inflammatory effects of quercetin: a review. Polymer Bull. 2023;80:241–62.

Article CAS Google Scholar

Chang Y, Lin H, Chan S, Yeh S. Effects of quercetin metabolites on the enhancing effect of b-carotene on DNA damage and cytochrome P1A1/2 expression in benzo [a] pyrene-exposed A549 cells. Food Chem. 2012;133:445–50.

Article CAS PubMed Google Scholar

Kim JY, Kim EH, Park SS, Lim JH, Kwon TK, et al. Quercetin sensitizes human hepatoma cells to TRAIL-induced apoptosis via Sp1-mediated DR5 upregulation and proteasome-mediated c-FLIPS down-regulation. J Cell Biochem. 2008;105:1386–98.

Article CAS PubMed Google Scholar

Yoshino S, Hara A, Sakakibara H, Kawabata K, Tokumura A, et al. Effect of quercetin and glucuronide metabolites on the monoamine oxidase-A reaction in mouse brain mitochondria. Nutrition. 2011;27:847–52.

Article CAS PubMed Google Scholar

Xue Y, Du M, Zhu MJ. Quercetin suppresses NLRP3 inflammasome activation in epithelial cells triggered by Escherichia coli O157:H7. Free Radic Biol Med. 2017;108:760–9.

Article CAS PubMed Google Scholar

Ishizawa K, Yoshizumi M, Kawai Y, Terao J, Kihira Y, Ikeda Y, et al. Pharmacology in health food: metabolism of quercetin in vivo and its protective effect against arteriosclerosis. J Pharmacol Sci. 2011;115:466–70.

Article CAS PubMed Google Scholar

Siriwong S, Teethaisong Y, Thumanu K, Dunkhunthod B, Eumkeb G. The synergy and mode of action of quercetin plus amoxicillin against amoxicillin-resistant Staphylococcus epidermidis. BMC Pharmacol Toxicol. 2016;17:1–14.

Article Google Scholar

Vipin C, Saptami K, Fida F, Mujeeburahiman M, Rao SS, Arun AB, et al. Potential synergistic activity of quercetin with antibiotics against multidrug-resistant clinical strains of Pseudomonas aeruginosa. PLoS ONE. 2020;15:e0241304.

Article CAS PubMed PubMed Central Google Scholar

Emiliano YS, Almeida-Amaral EE. Efficacy of apigenin and miltefosine combination therapy against experimental cutaneous leishmaniasis. J Nat Prod. 2018;81:1910–3.

Article CAS PubMed Google Scholar

Huffman AM, Ayariga JA, Napier A, Robertson BK, Abugri DA. Inhibition of Toxoplasma gondii growth by dihydroquinine and its mechanisms of action. Front Cell Infect Microbiol. 2022;12:852889. https://doi.org/10.3389/fcimb.2022.852889.

Article CAS PubMed PubMed Central Google Scholar

Blais J, Garneau V, Chamberland S. Inhibition of Toxoplasma gondii protein synthesis by azithromycin. Antimicrob Agents Chemother. 1993;37:1701–3.

Article CAS PubMed PubMed Central Google Scholar

Abugri DA, Witola WH, Russell AE, Troy RM. In vitro activity of the interaction between taxifolin (dihydroquercetin) and pyrimethamine against Toxoplasma gondii. Chem Biol Drug Des. 2018;91:194–201.

Article CAS PubMed Google Scholar

Abugri DA, Witola WH. Interaction of apigenin-7-O-glucoside with pyrimethamine against Toxoplasma gondii growth. J Parasit Dis. 2020;44:221–9.

Article PubMed Google Scholar

Ma CI, Tirtorahardjo JA, Jan S, Schweizer SS, Rosario SA, Du Y, et al. Auranofin resistance in Toxoplasma gondii decreases the accumulation of reactive oxygen species but does not target parasite thioredoxin reductase. Front Cell Infect Microbiol. 2021;11:20. https://doi.org/10.3389/fcimb.2021.618994.

Article CAS Google Scholar

Charvat RA, Arrizabalaga G. Oxidative stress generated during monensin treatment contributes to altered Toxoplasma gondii mitochondrial function. Sci Rep. 2016;6:1–17.

Article Google Scholar

Sharma HN, Catrett J, Nwokeocha OD, Boersma M, Miller ME, Napier A, et al. Anti-Toxoplasma gondii activity of Trametes versicolor (Turkey tail) mushroom extract. Sci Rep. 2023;13:8667.

Article CAS PubMed PubMed Central Google Scholar

Zhang JL, Si HF, Shang XF, Zhang XK, Li B, Zhou XZ, et al. New life for an old drug: in vitro and in vivo effects of the anthelmintic drug niclosamide against Toxoplasma gondii RH strain. Int J Parasitol Drugs Drug Resist. 2019;9:27–34.

Article CAS PubMed Google Scholar

Zhang J, Si H, Lv K, Qiu Y, Sun J, Bai Y, et al. Licarin-B exhibits activity against the Toxoplasma gondii RH strain by damaging mitochondria and activating autophagy. Front Cell Dev Biol. 2021;9:684393.

Article PubMed PubMed Central Google Scholar

Zhang J, Si H, Li B, Zhou X, Zhang J. Myrislignan exhibits activities against Toxoplasma gondii RH strain by triggering mitochondrial dysfunction. Front Microbiol. 2019;10:2152. https://doi.org/10.3389/fmicb.2019.02152.

Article PubMed PubMed Central Google Scholar

Syn G, Anderson D, Blackwell JM, Jamieson SE. Toxoplasma Gondii infection is associated with mitochondrial dysfunction in-Vitro. Front Cell Infection Microbiol. 2017;7:512. https://doi.org/10.3389/fcimb.2017.00512.

Article CAS Google Scholar

Spalenka J, Escotte-Binet S, Bakiri A, Hubert J, Renault J-H, Velard F, Duchateau S, Aubert D, Huguenin A, Villena I. Discovery of new inhibitors of Toxoplasma gondii via the pathogen box. Antimicrob Agents Chemother. 2018;62(2):10–1128.

Article Google Scholar

Sanford AG, Schulze TT, Potluri LP, Hemsley RM, Larson JJ, Judge AK, et al. Novel Toxoplasma gondii inhibitor chemotypes. Parasitol Int. 2018;67:107–11.

Article CAS PubMed PubMed Central Google Scholar

van der Ven AJ, Schoondermark-van de Ven EM, Camps W, Melchers WJ, Koopmans PP, van der Meer JW, et al. Anti-toxoplasma effect of pyrimethamine, trimethoprim and sulphonamides alone and in combination: implications for therapy. J Antimicrob Chemother. 1996;38:75–80.

Article PubMed Google Scholar

Derouin F, Chastang C. In vitro effects of folate inhibitors on Toxoplasma gondii. Antimicrob Agents Chemother. 1989;33:1753–9.

Article CAS PubMed PubMed Central Google Scholar

Goodman CD, Useglio M, Peirú S, Labadie GR, McFadden GI, Rodríguez E, et al. Chemobiosynthesis of new antimalarial macrolides. Antimicrob Agents Chemother. 2013;57:907–13.

Article CAS PubMed PubMed Central Google Scholar

Wilson DW, Goodman CD, Sleebs BE, Weiss GE, de Jong NW, Angrisano F, et al. Macrolides rapidly inhibit red blood cell invasion by the human malaria parasite Plasmodium falciparum. BMC Biol. 2015;13:1–19.

Article CAS Google Scholar

Liu Y, Kam W, Ding J, Sullivan D. Impact of azithromycin on lipid accumulation in immortalized human meibomian gland epithelial cells. Invest Ophthalmol Vis Sci. 2013;54(15):4317–4317.

Google Scholar

Fa N, Ronkart S, Schanck A, Deleu M, Gaigneaux A, Goormaghtigh E, Mingeot-Leclercq M-P. Effect of the antibiotic azithromycin on thermotropic behavior of DOPC or DPPC bilayers. Chem Phys lipids. 2006;144(1):108–16.

Article CAS PubMed Google Scholar

Lode H, Borner K, Koeppe P, Schaberg T. Azithromycin—review of key chemical, pharmacokinetic and microbiological features. J Antimicrobial Chemother. 1996;37(Suppl C):1–8.

Article CAS Google Scholar

Fonseca-Silva F, Inacio JD, Canto-Cavalheiro MM, Almeida-Amaral EE. Reactive oxygen species production and mitochondrial dysfunction contribute to quercetin induced death in Leishmania amazonensis. PLoS ONE. 2011;6:e14666.

Article CAS PubMed PubMed Central Google Scholar

Sifaoui I, Lopez-Arencibia A, Martín-Navarro CM, Ticona JC, Reyes-Batlle M, Mejri M, et al. In vitro effects of triterpenic acids from olive leaf extracts on the mitochondrial membrane potential of promastigote stage of Leishmania spp. Phytomedicine 2014;21(12):1689–1694.

Article CAS PubMed Google Scholar

Das K, Roychoudhury A. Reactive oxygen species (ROS) and response of antioxidants as ROS-scavengers during environmental stress in plants. Front Environ Sci. 2014;2:53.

Article Google Scholar

Das R, Roy A, Dutta N, Majumder HK. Reactive oxygen species and imbalance of calcium homeostasis contributes to curcumin induced programmed cell death in Leishmania donovani. Apoptosis. 2008;13:867–82.

Article CAS PubMed Google Scholar

Salehi B, Machin L, Monzote L, Sharifi-Rad J, Ezzat SM, Salem MA, et al. Therapeutic potential of quercetin: new insights and perspectives for human health. ACS Omega. 2020;5:11849–72.

Article CAS PubMed PubMed Central Google Scholar

Dajas F. Life or death: neuroprotective and anticancer effects of quercetin. J Ethnopharmacol. 2012;143:383–96.

Article CAS PubMed Google Scholar

Download references

The following reagents were obtained through the NIH Biodefense and Emerging Infections Research Resources Repository, NAID, NIH, and BEI Resources: Vero Kidney cells (African green monkey), expressing luciferase (Luc2p); NR-10385 MDCK kidney cells. The RH-(YFP)2 cell line was kindly provided by Prof. William H. Witola from the College of Veterinary Medicine at the University of Illinois, Urbana-Champaign, IL USA. We would like to thank Bernard BA Efa for transporting the plates for the microscopy work.

No external funding was provided. The work was supported by Alabama State University through the Department of Biological Sciences in the division of Microbiology PhD program.

Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, 36104, USA

Daniel A. Abugri, Sandani V. T. Wijerathne, Homa Nath Sharma, Joseph A. Ayariga, Audrey Napier & Boakai K. Robertson

Microbiology PhD Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, 36104, USA

Daniel A. Abugri, Sandani V. T. Wijerathne, Homa Nath Sharma & Boakai K. Robertson

Laboratory of Ethnomedicine, Parasitology and Drug Discovery, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, 36104, USA

Daniel A. Abugri & Homa Nath Sharma

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

You can also search for this author in PubMed Google Scholar

DAA conceived the information. SW, JAA, HNS and DAA conducted the experiments. JAA, HNS and DAA performed the statistics. DAA wrote the initial paper. JAA, AN and BKR contributed technical advice and resources, made suggestions for the experimental approaches and read through the manuscripts to make corrections.

Correspondence to Daniel A. Abugri.

The study does not involve any vertebrate animals.

All authors agreed on the content of the paper for publication.

There is no competing interest.

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and Permissions

Abugri, D.A., Wijerathne, S.V.T., Sharma, H.N. et al. Quercetin inhibits Toxoplasma gondii tachyzoite proliferation and acts synergically with azithromycin. Parasites Vectors 16, 261 (2023). https://doi.org/10.1186/s13071-023-05849-3

Download citation

Received: 09 February 2023

Accepted: 26 June 2023

Published: 03 August 2023

DOI: https://doi.org/10.1186/s13071-023-05849-3

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative