Next Article in Journal / Special Issue
Light, Dyes, and Action: Photodynamic Inactivation of Leishmania amazonensis Using Methylene Blue, New Methylene Blue, and Novel Ruthenium-Based Derivatives
Previous Article in Journal
Effects of Preventive Exposure to High Doses of Alpha-Lipoic Acid (ALA) on Testicular and Sperm Alterations Caused by Scrotal Heat Shock in Mice
Previous Article in Special Issue
Leishmania in Texas: A Contemporary One Health Scoping Review of Vectors, Reservoirs, and Human Health
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

From Dyes to Drugs? Selective Leishmanicidal Efficacy of Repositioned Methylene Blue and Its Derivatives in In Vitro Evaluation

by
Deyvison Rhuan Vasco-dos-Santos
1,*,
Juliana Almeida-Silva
1,
Ludmila Ferreira de Almeida Fiuza
2,
Natalia Vacani-Martins
1,
Zênis Novais da Rocha
3,
Maria de Nazaré Correia Soeiro
2,
Andrea Henriques-Pons
1,
Eduardo Caio Torres-Santos
4 and
Marcos André Vannier-Santos
1,*
1
Laboratório de Inovação em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz (LITEB—IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
2
Laboratório de Biologia Celular, Instituto Oswaldo Cruz (LBC—IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
3
Instituto de Química, Universidade Federal da Bahia (UFBA), Campus Universitário de Ondina, Salvador 40170-290, BA, Brazil
4
Laboratório de Bioquímica de Tripanossomatídeos, Instituto Oswaldo Cruz (LBqT—IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
*
Authors to whom correspondence should be addressed.
Biology 2025, 14(12), 1709; https://doi.org/10.3390/biology14121709
Submission received: 16 November 2024 / Revised: 25 December 2024 / Accepted: 3 January 2025 / Published: 30 November 2025

Simple Summary

Cutaneous leishmaniasis is an insect-borne disease caused by microscopic parasites called Leishmania, which enter the body cells and can cause sores, including on the face. Used treatments are mainly old, painful, and can harm the liver, kidneys, and heart. Therefore, it is important to discover novel effective, safe, and orally administered medications. In this study, we tested methylene blue, the first fully synthetic drug used in medicine. This vintage compound was previously used for blood disorders, including malaria, and is presently being evaluated for other conditions. Here, we approached the antiparasitic activity of methylene blue and its derivatives upon Leishmania and tested whether they affected laboratory-grown cells. We found that the substances effectively diminished parasite survival without exerting toxic effects on skin cells and with only low to moderate toxicity to liver and kidney cells. It is noteworthy that methylene blue can be protective for heart and nerve tissues. These data suggest that the tested substances are promising for the development of treatments for cutaneous leishmaniasis.

Abstract

Chemotherapy for cutaneous leishmaniasis is hindered by high toxicity, adverse effects, and increasing drug resistance. Thus, safer and more selective therapies are urgently required. Here, we evaluated the antiparasitic efficacy of methylene blue (MB) and new MB, as well as novel ruthenium complex derivatives (NMB-B and NMB-P) against promastigote and amastigote forms of Leishmania amazonensis. Their cytotoxicity and selectivity on L929, HepG2, VERO, J774.G8 cells, and murine peritoneal macrophages were measured. Mechanisms of action were explored via flow cytometry, assessing morphological changes, mitochondrial depolarization, ROS production, and cell death. The compounds inhibited parasite proliferation in a dose and time-dependent manner, achieving submicromolar efficacy against amastigotes (NMB-P = 0.46 μM). No cytotoxicity was observed on L929, J774.G8, and VERO cells (except NMB), while HepG2 and murine peritoneal macrophages showed low to moderate toxicity. Selective indexes reached 84 for promastigotes and over 500 for amastigotes. The compounds induced mitochondrial depolarization by up to 61% and a five-fold increase in ROS levels, leading to structural damage and parasite death via late apoptosis/necrosis-like mechanisms. These findings indicate that the compounds act selectively and trigger the release of oxidative species, exerting leishmanicidal activity and warranting further investigation.

1. Introduction

Leishmaniasis encompasses a group of tropical neglected diseases, also known as the diseases of poverty, being intrinsically linked to malnutrition and socioeconomic vulnerabilities, with an estimated one billion individuals living at a risk of infection [1,2,3,4]. These diseases are caused by the protozoa of the genus Leishmania, transmitted by phlebotomine sandflies [5,6]. Recent studies have also demonstrated the possibility of oral and intragastric transmission in experimental hamster infection [7]. Despite the spectral clinical manifestations in humans, there are three main forms of leishmaniasis: visceral, cutaneous, and mucocutaneous [8,9].
In the Americas, cutaneous leishmaniasis (CL) is the most prevalent form, caused by various species including, Leishmania amazonensis. The complications range from ulcers to immunopathogenic syndromes, potentially escalating to visceral leishmaniasis depending on the parasite virulence and the patient’s immunocompetence [10,11,12]. CL can also result in permanent and disfiguring lesions, leading to social stigmatization, psychological distress, and decreased work capacity and quality of life [13,14,15].
The therapeutic arsenal for leishmaniasis remains limited, with pentavalent antimonials such as meglumine antimoniate (Glucantime®) and sodium stibogluconate (Pentostam®) being the primary choice for nearly seven decades [16,17,18]. These medications are associated with severe adverse effects, including myalgia, arthralgia, pancreatitis, and gastrointestinal and respiratory disorders [19,20]. They are also cardio [21,22,23], hepato- [24], geno- [25], neuro- [26,27], and nephrotoxic [28,29], making them contraindicated for groups such as pregnant [30] and renal patients [31].
Additionally, the administration of antimonials via parenteral routes over extended periods is associated with significant pain, hospitalization requirements, reduced patient adherence, and treatment continuity, leading to therapeutic failures [32,33,34] and resistance phenotype selection [35]. Hence, there is a pressing need for the development of new therapies that are not only accessible and well tolerated but also can be administered orally or topically and require brief treatment courses [36,37].
Drug repositioning emerges as an advantageous strategy among the available alternatives. This approach utilizes existing drugs with well-established pharmacokinetic and safety profiles, effectively reducing development time, costs, and associated risks. As a result, it potentially shortens the timeline from drug development to patient access, ensuring the faster availability of treatments, particularly for those relying on public healthcare systems [38,39]. Examples of repurposed drugs for testing in Leishmania species include methylene blue (MB), the first synthetic substance used as a medication [40]. This vintage drug is still approached for distinct disorders and may be repositioned for conditions such as Alzheimer’s disease [41], acute liver failure [42], and COVID-19 [43].
Considering the aforementioned cardio- and neurotoxicity of antileishmanial drugs, which may be cumulative [44], it is important to point out that MB can display neuro- and cardioprotective effects [45]. The neuroprotection is relevant not only for the therapy that often requires multiple treatment courses, but also because Leishmania spp. infection is repeatedly associated with neurological manifestations in humans [46,47] and other animals [48], including perineural inflammation [49]. In this regard, it is interesting that MB is anti-inflammatory [50], able to inhibit neuroinflammation [51].
MB presents low cost, high hydrosolubility [52], and has been applied as an antimalarial for over a century [53]. Its antileishmanial action is associated with its potential as a photosensitizer in photodynamic therapy (PDT) [54,55,56], inducing mitochondrial damage [57], with effects observed in CL caused by L. amazonensis in experimental murine [58] and hamster [59] infections as well as in human cases [60,61]. However, studies elucidating the antileishmanial activity of MB without photostimulation are incipient.
Thus, we evaluated the in vitro activity of MB, new methylene blue (NMB), and its ruthenium complexes (new methylene blue B—NMB-B and new methylene blue P—NMB-P) against promastigote and ex vivo amastigote forms of L. amazonensis, without light exposure. Ruthenium complexes have been described as catalysts of potent and synergistic leishmanicidal activities [62,63,64,65], encouraging the investigation of NMB-B and NMB-P complexes.
In this study, we also investigated the cytotoxicity and selectivity of the compounds in different mammalian cells. Evidence suggests that the screening of drug candidates considering pharmacokinetics and pharmacodynamics is more effective when performed in multiple host cell types [66]. We have previously noticed that another ruthenium complex was fungicidal and triggered lipid peroxidation, damaging the stable cell walls of fluconazole-resistant Candida tropicalis, even with no photostimulation [67]. Therefore, we hypothesize that the compounds tested here can display promising leishmanicidal activity, even without photodynamic dynamization.

2. Material and Methods

2.1. Compounds and Reagents

MB, NMB (Figure 1A,B), and miltefosine (MT) were acquired from Sigma-Aldrich (St. Louis, MO, USA), as well as ruthenium trichloride (RuCl3·nH2O), 2,2′-bipyridine (bpy), and 1,10-phenanthroline (phen), which were utilized in the synthesis of NMB-B and NMB-P complexes (Figure 1C,D). These complexes were synthesized by the Instituto de Química da Universidade Federal da Bahia (UFBA—Salvador, BA, Brazil). MT was dissolved in sterile water and the other compounds in phosphate-buffered saline (PBS; Gibco—Grand Island, NY, USA). All substances were stored at −20 °C until use.
Schneider’s Insect Medium and Dulbecco’s Modified Eagle Medium (DMEM), penicillin/streptomycin (Pen/Str), L-glutamine, carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone (FCCP), Antimycin A (AA), RPMI 1640 without phenol, and brewer’s thioglycolate were obtained from Sigma-Aldrich (St. Louis, MO, USA). Fetal bovine serum (FBS) from Cultilab (Campinas, SP, Brazil) and Alamar Blue (AB) from Invitrogen (Eugene, OR, USA). FITC Annexin V Apoptosis detection Kit I and was purchased from BD Pharmingen (Franklin Lakes, NJ, USA); 7-Amino-Actinomycin D (7-AAD) from BioLegend (San Diego, CA, USA); tetramethylrhodamine ethyl (TMRE) from Molecular Probes (Carlsbad, CA, USA); and 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA) from PROC9 (Canoas, RS, Brazil).

2.2. Synthesis of Ruthenium Complexes

The target complexes were synthesized starting with RuCl3·nH2O as the primary material. The complexes cis-[RuCl2(X-Y)2] (where X-Y represents polypyridyl ligands such as phen and bpy) and cis-[Ru(NO2)2(X-Y)2](PF6)2 were intermediates in the preparation of the final complexes, cis-[Ru(NO)(NMB)(X-Y)2](PF6)4. To prepare these complexes, 0.05 g (0.094 mmol) of [RuCl2(X-Y)2] was dissolved in a 5 mL/5 mL ethanol/water mixture under an argon atmosphere and heated at 55 °C for 15 min. Subsequently, 0.035 g (~0.094 mmol) of NMB and 0.033 g (0.0187 mmol) of potassium nitrate were added. For all systems, the mixture was maintained under stirring and argon atmosphere at 55 °C for 2 h, followed by the addition of 100 mg of NH4PF6 (0.61 mmol) and 1 mL of HPF6 0.1 mol L−1 solutions. The resulting green precipitation was collected through filtration.

2.3. Ethical and Animal Statements

Access to genetic resources was granted by the National System for Genetic Heritage and Associated Traditional Knowledge (Sistema Nacional de Gestão do Patrimônio Genético e do Conhecimento Tradicional Associado—SisGen) under registration AD1C828. All animal study protocols were approved by the Comitê de Ética para o Uso de Animais (CEUA—IOC/Fiocruz) in Rio de Janeiro (lic. L038-2017 A4) and comply with the guidelines of the National Council for the Control of Animal Experimentation (Conselho Nacional de Controle de Experimentação Animal—CONCEA).

2.4. Culture of Parasitic Cells

Promastigote forms of Leishmania amazonensis (IFLA/BR/1967/PH8) were obtained from the Instituto Oswaldo Cruz Leishmania collection (CLIOC/Fiocruz). These cultures were maintained in Schneider’s Insect Medium, supplemented with 20% FBS and 1 μL/mL of Pen/Str at 26 °C. In vitro passages were conducted weekly, with promastigotes harvested during the late logarithmic phase for experiments on the 5th day of growth [68] (Figure S1A).
Ex vivo amastigote forms of L. amazonensis (MHOM/BR/77/LTB0016) were harvested and purified from lesions of male BALB/c mice. For this, a suspension containing 106 amastigotes was inoculated subcutaneously into the intraplantar region of the hind paw. After 30 days, the animals were euthanized, and their paws were excised and rinsed in 70% ethanol. The epidermis and necrotic tissues were mechanically dissociated. Subsequently, the paws were placed in Petri dishes containing RPMI 1640 medium supplemented with 5% FBS and 2% Pen/Str. To liberate the amastigotes, the tissues were scraped, and the suspension was transferred to conical tubes, vigorously homogenized using a 20 mL syringe, and centrifuged at 101× g for 15 min. The supernatant was transferred to another tube, homogenized with a syringe, and centrifuged at 764× g for another 15 min. The supernatant was discarded, and the pellet was resuspended in RPMI 1640 medium [69].

2.5. Culture of Mammalian Host Cells

HepG2 cells (human hepatocellular carcinoma), VERO cells (African green monkey kidney), and J774.G8 macrophages (murine reticulosarcoma) were maintained in DMEM medium, supplemented with 10% FBS, 1% Pen/Str at 37 °C and 5% CO2 [70,71]. For the cultivation of L929 fibroblasts (murine fibrosarcoma), RPMI 1640 medium without phenol red, supplemented with 2 mM L-glutamine, 10% FBS, and 1% Pen/Str, was used and maintained under the same conditions of temperature and CO2 as above [72].
Murine peritoneal macrophages—MPMs (primary cells)—were isolated from male Swiss Webster mice stimulated with 1 mL of 3% Brewer’s thioglycollate medium, administered intraperitoneally. After 96 h, the animals were euthanized, and the MPMs were collected by peritoneal lavage with approximately 10 mL of RPMI 1640 medium. The cell suspension was centrifuged at 382× g for 10 min, and the pellet was resuspended in medium supplemented with 5% FBS, 1% Pen/Str, and 2 mM L-glutamine. All cell cultures were maintained at 37 °C and 5% CO2 [73]. For the experiments, all cell types were cultured until confluency.

2.6. Leishmanicidal Activity Assay: Evaluation of Promastigotes and Ex Vivo Amastigotes

Promastigote and ex vivo amastigote forms of L. amazonensis (106 parasites/mL) were seeded into 96-well flat-bottom microplates and treated with various concentrations of the compounds, serially diluted in Schneider/RPMI 1640 medium to a final volume of 100 µL/well. Promastigotes were kept at 26 °C, whereas amastigotes were kept at 32 °C. After 24 h, parasite viability was assessed using the AB colorimetric method, and the data were analyzed using a spectrophotometer as previously described [74]. The results were used to determine the 50% Maximal Inhibitory Concentration values (IC50). For promastigote forms, viability was also spectrophotometrically evaluated using AB through growth curves in the presence of the compounds at IC50 and different time points (24–168 h). Wells with medium and without parasites were used as blanks, and wells with medium and untreated parasites were used as negative controls. MT was employed as the reference drug.

2.7. Cytotoxicity Assays on Different Mammalian Cell Types and Selective Indexes

J774.G8, VERO (105 cells/well), HepG2, L929 (0.5 × 105 cells/well) cells, and MPMs (3 × 105 cells/well) were centrifuged, resuspended in medium, plated in 96-well microplates, and incubated for 24 h under cultivation conditions. Subsequently, they were exposed to different concentrations of the compounds, serially diluted in medium to a final volume of 100 µL/well, and incubated for 24 h at 37 °C and 5% CO2. Cell viability was then assessed using the AB, and data were acquired by spectrophotometry [74]. The results were used to calculate the 50% Maximal Cytotoxicity Concentration (CC50). Untreated cells were used as negative controls, and cells exposed to the reference compound (MT) were used as positive controls. The Selectivity Index (SI) was calculated using the ratio SI = CC50 (mammalian cells)/IC50 (promastigote or amastigote forms of L. amazonensis).

2.8. Evaluation of Mechanisms of Action by Flow Cytometry

Initially, we treated promastigote forms of L. amazonensis (106 parasites/mL) with 200 μM of MB, the highest concentration used in the inhibition assays, and with the IC50 of each compound. We observed that the samples did not emit fluorescence in the channels of interest (FITC, PE, and PerCP—Figure S2). Next, both promastigotes and ex vivo amastigotes were exposed to the IC50 of the compounds for 24 h. They were then evaluated for mitochondrial membrane potential (ΔΨm), reactive oxygen species (ROS) production (5 × 106 parasites/mL), and detection of apoptosis/necrosis (105 parasites/mL). Data acquisition was performed using a Cytoflex S flow cytometer (Beckman Coulter, Brea, CA, USA) at the Flow Cytometry Facility—Unity of Multiparametric Analysis of Instituto Oswaldo Cruz—and analyzed using the Cytexpert software (version 2.5). In total, 10,000 events were acquired in the regions corresponding to L. amazonensis, previously established.

2.8.1. Determination of Mitochondrial Membrane Potential—ΔΨm

The parasites were incubated with 0.05 µM of TMRE at 28 °C for 30 min. Untreated parasites, labeled with 10 µM of FCCP and TMRE+, were used as positive controls. Untreated parasites labeled with TMRE were used as negative controls. TMRE labeling was evaluated by median fluorescence intensity (MFI) in the PE channel, from which the variation index of mitochondrial membrane potential (VIΔΨm) was calculated using the equation: VIΔΨm = (MT − MC)/MC, where MT corresponds to the MFI of TMRE in treated parasites, and MC corresponds to the MFI of TMRE in control parasites. Negative results indicate mitochondrial membrane depolarization.

2.8.2. Generation of Reactive Oxygen Species—ROS

For ROS production analysis, both parasite forms studied were labeled with 20 µM of H2DCFDA for 20 min at 28 °C in the dark. As a positive control, parasites were incubated with 10 µM of AA, while untreated parasites served as the negative control. The percentage of H2DCFDA labeling was measured in the FITC channel to calculate the variation index of ROS production (VIROS) using the equation: VIROS = MT/MC, where MT corresponds to the H2DCFDA MFI of treated parasites, and MC corresponds to the H2DCFDA MFI of control parasites.

2.8.3. Cell Death: Phosphatidylserine Exposure and Plasma Membrane Integrity

Cells were incubated with the FITC Annexin V Apoptosis Detection Kit I to detect apoptosis and necrosis. Additionally, for ex vivo amastigotes, labeling with pre-titrated 7-AAD probe was employed. All markers were used according to the manufacturers’ recommendations. Parasites heated to 60 °C for 15 min were used as positive controls, while untreated parasites were used as negative controls (Figure S3). The percentages of apoptotic and/or necrotic events were evaluated based on Annexin V (AV), propidium iodide (PI), and 7-AAD positivity in the FITC, PE, and PerCP channels, respectively.

2.8.4. Image Flow Cytometry

Parasites treated as described above for the apoptosis/necrosis detection assay and labeled with Annexin FITC/PI were analyzed by image flow cytometry. Sample acquisition was performed using the Amnis® ImageStream®X Mk II (Cytek® Biosciences Inc., Fremont, CA, USA) and analyzed with IDEAS software (version 6.3). Samples were acquired with a typical setup, including a 40× objective and a numerical aperture 0.75. Detection was performed using side scatter, brightfield, and two fluorescence images. The flow cytometer was equipped with a 488 nm excitation laser, and band pass filters of 480–560 nm and 595–642 nm were used to evaluate Annexin FITC and PI labeling, respectively. The image size of the cytometer was 0.5 × 0.5 μm per pixel, with a field of view of 60 × 128 μm at an image generation rate of 500 cells per second. Single positive parasites (Annexin FITC+ or PI+) were used to create the compensation matrix and generate the images.

2.9. Statistical Analysis

All assays were performed at least twice in three independent replicates. Data obtained were plotted using Microsoft Excel (version 2406), and GraphPad Prism® version 8 (GraphPad Software Inc., San Diego, CA, USA) was used to calculate IC50, CC50, and perform statistical analyses. One-way analysis of variance (ANOVA) followed by Dunnett’s post-test was used to compare groups. Differences were considered significant when p < 0.05 (*), p < 0.001 (**), and p < 0.0001 (***) as compared to the controls.

3. Results

3.1. Compounds Surpass the Efficacy of the Reference Drug in Inhibiting Different Forms of Leishmania amazonensis

The inhibitory actions of MB, NMB, NMB-B, and NMB-P were evaluated on promastigote and ex vivo amastigote forms of L. amazonensis. All compounds reduced the viability of both parasitic forms in a concentration-dependent manner (Figure 2A1–B5) and significantly (p < 0.05) as compared to the negative control. Some compounds were more effective at lower concentrations than the reference drug, MT. In promastigotes, at 1.56 μM and 3.12 μM, all compounds caused greater antiparasitic effects than MT (Figure 2C,D). In ex vivo amastigotes, NMB, NMB-B, and NMB-P showed more inhibitory action at doses < 0.10 μM, while at concentrations below 0.28 μM, both NMB and NMB-P surpassed the efficacy of MT (Figure 2E,F). IC50 values were obtained from dose–response assays (Table 1).
NMB and ruthenium complexes stood out for promastigote forms with IC50 < 5.50 μM, especially NMB-P with an IC50 value of 2.84 ± 0.80 μM. These results were highly significant (p < 0.0001) compared to MT. Conversely, MB showed low leishmanicidal activity upon this developmental form (IC50 = 61.44 ± 4.41 μM). However, ex vivo amastigote forms were more susceptible to all tested compounds, with IC50 values reduced by 2- to 6-fold compared to promastigotes. NMB-P achieved submicromolar IC50 values (0.46 ± 0.34 μM), and other treatments demonstrated promising results.
Based on IC50 determinations (Table 1), we evaluated the survival and proliferation of promastigote forms incubated for 24 to 168 h (Figure 3). The growth curves showed highly significant (p < 0.0001) and time-dependent inhibition compared to the control at all periods analyzed (Figure 3A). Miltefosine showed more pronounced growth peaks, whereas tested compounds showed a marked pronounced and progressive decline, especially with MB, which caused total inhibition from 96 h (Figure 3B).
These data corroborate preliminary results (Figure S1B1–B4), where treating promastigotes with different concentrations of MB (3.13–200 μM) showed a significant reduction (p < 0.05) at 48 h at higher concentrations (from 50 μM) and total viability loss from 12.50 μM at 72 h. The growth profiles of NMB, NMB-B, and NMB-P are like the reference drug (Figure 3C,E). A significant reduction (p < 0.05) in proliferation from 24 h was observed in MB from 48 h with NMB-B; from 72 h with NMB; and 96 h with NMB-P compared to MT. At 144 h, NMB-B showed a highly significant effect (p < 0.0001), while MB maintained this efficacy between 96 and 168 h.

3.2. Cytotoxicity on Different Mammalian Cell Types: Selective Efficacy of Repositioned and Novel Compounds

The cytotoxicity of MB, NMB, NMB-B, and NMB-P was evaluated in five different types of mammalian host cells, four cell lines (L929, HepG2, VERO, and J774.G8), and one primary cell type (MPMs). The compound’s toxicity was compared with MT, a widely known leishmanicidal drug. Dose–response curves (Figure 4) demonstrated that the reduction in viability was dose-dependent in all treatments. MB, at lower concentrations (37–100 μM), resulted in viability > 50% (Figure 4A1,B1,C1,D1), except on MPMs. Both NMB and NMB-B maintained this viability pattern in L929 cells (Figure 4A2,A3). MT, at concentrations ≥ 300 μM, eliminated the viability of VERO cells (Figure 4C5), whereas NMB-P was less toxic to these cells (Figure 4C4). The ruthenium complexes exhibited similar action to MT on MPMs, with viability > 50% at exposures up to 70 μM (Figure 4E3–E5).
Subsequently, the CC50 and respective SI for promastigote forms (SIP) and ex vivo amastigotes (SIA) were determined (Table 1). The compounds exhibited low to moderate or no toxicity, with CC50 values up to 521 times higher than the IC50 observed for L. amazonensis. Among the 40 conditions analyzed, 45% exhibited SI between 10 and 50, 15% between 50 and 100, and 13% exceeded 100. MB showed the best activity on L929 cells (CC50 = 321.65 ± 3.74 μM, SIP = 5.24 and SIA = 20.55), as did NMB (CC50 = 280.25 ± 2.19 μM) with an SIA close to that of MT (SIA NMB = 105.35 and SIA MT = 116.20). The ruthenium complexes were less toxic than the reference drug on L929 and VERO cells.
Concerning macrophages (J774.G8 and MPMs), which are targeted host cells of Leishmania infection, J774.G8 cells were less susceptible than MPMs, including treatment with MT. However, regarding selectivity, the SIP and SIA with NMB-P were higher in MPMs than in J774.G8. In contrast, with MT, the SI values for both parasite forms were lower in MPMs compared to lineage-derived macrophages.

3.3. Leishmania amazonensis ΔΨm Reduction

Flow cytometry was employed on parasites exposed for 24 h to the IC50 of the compounds to evaluate the possible mechanisms of action. Initially, ΔΨm was assessed using TMRE labeling in promastigote (Figure 5A1–A7) and ex vivo amastigote forms (Figure 5B1–B7) of L. amazonensis. Morphology dot plots (SSC × FSC—Figure 5A1,B1) showed two distinct populations based on relative cell size and granularity, defined as parasites with lower (LP—Low Population) and higher viability (HP—High Population), presented in gray and blue, respectively (Figure 5). This profile was confirmed by the AV/PI viability assay (Figure S4). Untreated parasites exhibited higher fluorescence when incubated with TMRE alone than those coincubated with FCCP (overlays in Figure 5A2,B2). Furthermore, controls showed high enrichment in HP (promastigotes = 94.73 ± 3.37% and amastigotes = 67.48 ± 8.50%—Figure 5A3,B3), whereas treated parasites exhibited reductions (up to 25%) in this subpopulation (Figure 5A4–A7,B4–B7).
The VIΔΨm are presented in Table 2. In LP promastigotes, the compounds, particularly MB and NMB-P (p < 0.05—Figure 5C), reduced 13% to 41%. Mitochondrial damage was greater in amastigotes, except for the treatment with NMB-P. In LP ex vivo amastigotes, the loss of potential ranged from 27% to 37%, while in HP, it was higher, varying from 43% to 61%. Considering both subpopulations, the data in amastigotes are notable, with decreases of 98%, 90%, and 76% after treatment with MB, NMB-B, and NMB, respectively. Therefore, these results indicate that the compounds significantly impact the ΔΨm of L. amazonensis in a manner dependent on the compound and parasitic form.

3.4. ROS Production Triggering

In the analysis of ROS generation using the H2DCFDA probe, AA was used as an inducer and control for the technique (overlays in Figure 6A2,B2). Untreated promastigotes exhibited fluorescence predominantly in the HP region, with 97.23 ± 2.44% of events (Figure 6A3). In contrast, treated parasites showed enrichment in the LP region, ranging from 7.00 ± 0.86% to 14.32 ± 3.10% (Figure 6A4–A7), indicating cellular damage. This profile was not observed in ex vivo amastigotes, where the percentages were similar to the control (Figure 6B3–B7). In this parasite form, the VIROS of all compounds were similar to AA in both LP and HP (Table 3Figure 6D). Conversely, in promastigotes, there was a remarkable increase in VIROS. MB and NMB tripled the ROS generation (p < 0.05), while NMB-B and NMB-P enhanced it five-fold (p < 0.001), surpassing AA (VIROS = 2.07 ± 0.23—Figure 6C). In the LP region, MB and NMB exceeded AA (VIROS = 0.52 ± 0.13), while NMB-B and NMB-P doubled this value.

3.5. Cellular Death in Leishmania amazonensis: Potential Mechanisms from Early Apoptosis to Necrosis

After 24 h of treatment with the IC50 of the compounds and labeling with AV/PI, mortality in promastigote forms ranged from 38.93 ± 8.06% to 57.10 ± 9.03% (Figure 7), confirming the inhibition data (Figure 2A1–A5). MB and NMB-B were notably significant (p < 0.05—Figure 7J). Approximately 12 ± 1.62% of events were AV+ and PI, while 25 ± 6.37% were PI+, suggesting late apoptosis or necrosis. Preliminary assays showed a drastic reduction in viability from 99.81% to 12.89% with 200 μM of MB (Figure S5).
According to the analysis of subpopulations (Figure S4), the LP region showed more apoptotic and/or necrotic events compared to the HP region, reflecting the greater sensitivity of parasites in this area to the compounds. All compounds in the LP region exhibited significantly higher AV+ events (p < 0.05) than the control. Even in the more resistant HP region, there was a significant increase in AV+ events with MB and NMB-B (p < 0.05). Additionally, exposure to NMB-P in the HP region resulted in a significantly (p < 0.0001) higher number of necrotic events, consistent with its IC50 (Table 1).
Image cytometry provided insights into the cellular death processes (Figure 8). Untreated parasites (Figure 8A1) displayed elongated and flagellated morphology typical of promastigotes, with high refringence and intact membranes. The absence of AV and PI labeling confirms cellular integrity (Figure 8A2–A4). However, the treatments resulted in evident structural damage (Figure 8B1,C1,D1,E1), possibly associated with different types of cell death, such as autophagy, apoptosis, and necrosis. Observed damages included cell rounding, cytoplasmic degradation, reduced refringence, membrane damage, flagellum loss, and possible vacuolation. The green fluorescence indicating phosphatidylserine exposure suggests early apoptosis (Figure 8B2,C2,D2,E2), while red fluorescence (PI binding to DNA) indicated late apoptosis or necrosis, with possible DNA fragmentation (Figure 8C3,C4) and release of genetic material into the extracellular medium (Figure 9B4,D4,E4).
High phosphatidylserine exposure was detected in ex vivo amastigote controls with AV labeling (77.13%), making it unsuitable as a cell death marker (Figure S6C), consistent with findings by Balanco et al. [75] and Wanderley et al. [76]. This pattern was absent in promastigotes [77,78]. Image cytometry showed control parasites with typical oval shape, non-prominent flagellum, intact membranes, and high refringence (indicative of parasite viability) while being AV+ (Figure S6E). Consequently, 7-AAD staining was performed (Figure 9). All compounds induced significant cell death (p < 0.05), ranging from 23.55 ± 2.56% to 65.42 ± 2.92%, suggestive of late apoptosis/necrosis. MB demonstrated the highest mortality rate (Figure 9C) with strong significance (p < 0.0001), followed by NMB and NMB-B (Figure 9D,E).
Subpopulation analysis (Figure S7) revealed that approximately 75.02 ± 0.57% of events in controls were in the HP region, with only 6 ± 2.08% of parasites being 7-AAD+, indicating high viability. Treatment significantly increased positivity (p < 0.001) from 19.60 ± 1.39% to 82.74 ± 3.95%, indicating membrane disruption and necrotic cell death. In the LP region, which corresponds to 17.71 ± 1.51% to 33.51 ± 3.12% of total events, 7-AAD+ percentages ranged from 0.42 ± 0.17% to 2.24 ± 0.67%. This lower labeling in LP might be due to the higher susceptibility of ex vivo amastigote forms to treatment (Table 1) and the LP region representing parasites with lower viability. Thus, the low 7-AAD labeling might be associated with secondary necrosis, where extensive cellular damage reduces the availability of detectable genetic material.

4. Discussion

The pursuit of more selective and less toxic therapies for leishmaniasis, particularly those administered orally and designed to overcome the parasite’s adaptive strategies and drug resistance mechanisms, remains a significant obstacle. This challenge can be interpreted as a “coevolutionary arms race” considering the Red Queen hypothesis [79]. In addition, the high toxicity and costs have largely hindered chemotherapy. Thus, the discovery of new drugs for CL capable of overcoming current obstacles is an urgent demand.
Numerous research teams have focused on evaluating substances with activity against L. amazonensis over the last decade exploring the activity of natural products [80,81,82,83,84,85,86], novel chemotypes [87,88,89,90,91], and repurposed drugs [92], using 24-h kinetics with IC50 values expressed in µM, as conducted in this study. These works emphasize in vitro investigations of mechanisms of action in promastigotes targeting the membrane and mitochondria, alongside cytotoxicity and selectivity analyses in mammalian cells.
Our findings align with literature, particularly in exploring mechanisms targeting different parasite structures, and in cytotoxicity and selectivity assessments. Notably, IC50 values ≤ 5.00 µM were observed in promastigotes using NMB, NMB-B, and NMB-P, as well as submicromolar concentration in amastigotes, a highly relevant parasite form for treatment (Table 1). There is evidence suggesting sensitivity similarities between ex vivo and intracellular amastigotes, warranting further investigation. Present et al. [93] evaluated a tubercidin analog and MT in ex vivo and intracellular amastigotes, reporting a mere 0.15 µM difference for the analog and 1.66 µM for MT. Similarly, Santos et al. [69] observed approximately a 4 µM difference between these amastigote populations challenged with nitroaromatic compounds.
In vitro potency determination is important for screening drug candidates for infectious diseases and hit-to-lead profiles serve as guidelines. While reference values are established for visceral leishmaniasis (VL) [94,95], data for CL remain scarce despite its substantial impact [37]. Katsuno et al. [96] proposed a generic criterion for substance selection, suggesting an SI > 10 in mammalian cells like HepG2 or VERO. For VL, they recommend an IC50 < 10 µM and an SI > 100. Our data are consistent with these guidelines, with 75% of compounds tested in promastigotes and amastigotes achieving an IC50 < 10 µM. Additionally, upon testing the cytotoxicity of four compounds across two parasitic forms and five cell types (HepG2, Vero, L929, J774.G8, and MPMs), we obtained SI values > 10 in most conditions.
Among the various cells susceptible to Leishmania infection, macrophages play a critical role in differentiation, proliferation, and infection regulation [97,98,99,100]. Previous studies [81,82,83,84,85,86,87,88,89] reported the greater sensitivity of primary cells, corroborating our findings, including for MT (Table 1). NMB-P stood out with SI > 150 on ex vivo amastigotes for primary and lineage macrophages. Preliminary in vitro screening with mammalian cell lines, especially hepatic and renal models, is important for the early identification of promising leishmanicidal compounds, given the well-known toxicity associated with first-line medications. However, further studies are needed to assess hepato- and nephrotoxicity in complex systems. In leishmaniasis chemotherapy, these toxic effects are not exclusive to antimonials [31,101,102,103] but have also been reported for amphotericin B [104,105,106,107,108,109], paromomycin [110], pentamidine [111,112], and MT [113,114,115,116].
In the cytotoxicity analysis using HepG2 cells, a widely used hepatic tumor line including studies on compound metabolism [117,118], MB, and NMB exhibited low toxicity (CC50 > 70 μM). These results contrast with those of Hameed et al. [119], who evaluated 101 substances against parasites, with 64.35% targeting Leishmania mexicana and found a correlation between high activity in trypanosomatids and elevated cytotoxicity in HepG2. In our investigation, only NMB-B and NMB-P demonstrated moderate cytotoxicity (CC50 < 36 μM) at concentrations similar to Amphotericin B (35.24 μM), as reported by Araújo et al. [120].
In VERO cells, the compounds exhibit antileishmanial activity at non-cytotoxic concentrations, a critical criterion for advancing drug candidates [121]. NMB-B and NMB-P demonstrated lower toxicity (CC50 > 230 μM) compared to MT (CC50 = 168.05 μM), with superior SI (Table 1). A previous study supports our findings, showing that phenothiazine possesses low or negligible cytotoxicity across various cell lines, underscoring their potential in leishmaniasis chemotherapy [122].
The role of non-canonical cells, such as fibroblasts, in leishmaniasis has been underestimated despite their ability to sustain parasite multiplication and survival, potentially contributing to persistence after self-healing or treatment [66]. The possibility of Leishmania remaining dormant in fibroblasts requires new therapeutic approaches. Cavalcante-Costa et al. [123] demonstrated that L. amazonensis infects fibroblasts through mechanisms distinct from phagocytosis, subverting cellular functions such as Ca2+ signaling and lysosomal exocytosis. Recently, Yektaeian et al. [124] revealed the potential for parasite detection in fibroblasts using iron oxide nanoparticles. Given their therapeutic importance, leishmanicidal substances have been evaluated for toxicity in hFB [125], 3T3 [126,127,128], and L929 [129,130,131] fibroblasts. In this study, the compounds showed no significant toxicity in L929 cells (CC50 > 150 µM).
Regarding the tested compounds, for MB without light treatment, the IC50 for promastigotes was reported as 100 μM [60], with no cytotoxicity observed in fibroblasts and macrophages [132], similar to our results for both MB and NMB on L929 and J744.G8 (Table 1). When combined with PDT, MB shows efficacy against species causing CL, promoting healing and reducing parasite burden in vivo [58,133,134,135] and in vitro, including against MT-resistant strains, with damage associated with ROS induction and ΔΨm loss [56,136]. Cabral et al. [136] reported a 2-fold increase in ROS levels and a 60% reduction in ΔΨm in L. amazonensis after MB treatment with PDT.
Aureliano et al. [57] demonstrated that PDT enhances MB activity, leading to mitochondrial damage observed through electron microscopy, an approach that may be instrumental in the elucidation of the antiparasitic agent mechanism of action [137]. However, the electron micrographs revealed vacuoles containing electron-dense material, which were not explored in the study and had been previously described [138,139]. These vacuoles may correspond to empty acidocalcisomes, previously reported in Leishmania major as compartments related to lysosomes, originating from multivesicular bodies [140]. Moreover, the authors did not discuss the presence of necrotic cells and the marked induction of autophagy, a process extensively studied [141], including Leishmania [142].
We present similar findings, even without PDT, where both MB and NMB increased ROS generation up to 5-fold (Table 3) and reduced ΔΨm by up to 61% (Table 2). These mechanisms are linked to the high mortality observed in promastigotes (Figure 7) and ex vivo amastigotes (Figure 9) following treatment with the compounds’ IC50. Significant changes in mitochondrial activity and cell morphology, including cell body rounding, were also reported in L. major and Leishmania braziliensis under PDT with MB by Pinto et al. [52], as documented through image cytometry for MB, NMB, and its complexes (Figure 8B1,C1,D1,E1).
Ruthenium complexes (RCs) have demonstrated potent antileishmanial activity in vitro, achieving inhibitory concentrations at low to submicromolar doses [64,143]. Costa et al. [63] observed IC50 values ranging from 0.52 to 7.52 µM in L. amazonensis promastigotes, though with cytotoxicity in Raw 264.7 macrophages (CC50 between 2.14 and 8.73 µM). Our data confirm the efficacy of RCs with NMB-B (IC50pro = 5.48 µM; IC50ama = 3.16 µM) and NMB-P (IC50pro = 2.84 µM; IC50ama = 0.46 µM), as well as their low cytotoxicity in J774.G8 macrophages (CC50 > 65 µM) and high selectivity against both parasite forms (SI > 10). These results are in agreement with Fandzloch et al. [144], who reported CC50 > 70 µM on J774.2 cells and > 58 µM on VERO cells. In these cell types, we observed CC50 > 238 µM and SI ranging from 45 to 525.30. In vivo, RCs significantly reduced lesion size and parasite load [65,145].
One of the mechanisms of action of RCs involves the release of nitric oxide (NO), which plays a crucial role in controlling Leishmania infections by inducing apoptotic-like death in intracellular amastigotes [146,147]. In promastigotes, RCs significantly reduced proliferation, while in infected macrophages, parasitic death was induced by increased NO production, concomitant with elevated expression of Akt, NF-kB, and iNOS in the macrophages [148]. In environments with ROS, NO can react to form more reactive and toxic species [149].
Costa et al. [150] suggest that RCs act as oxidizing agents, inducing L. amazonensis death through an apoptosis-like process characterized by increased ROS production and ΔΨm depolarization, consistent with our findings (Table 2 and Table 3). Additionally, similar morphological alterations were observed, such as cell rounding, mitochondrial compromise, and DNA fragmentation, also described in L. major [62], suggesting a cell death profile through a mechanism resembling late apoptosis/necrosis, as demonstrated by the markers PI and 7-AAD (Figure 7, Figure 8 and Figure 9).

5. Conclusions

Overall, the compounds demonstrated promising leishmanicidal activity against both stages of L. amazonensis, achieving submicromolar concentration and showing selectivity toward renal cells, hepatocytes, fibroblasts, and macrophages, thereby confirming our initial hypothesis. Additionally, the substances exhibited early/late apoptosis features, such as phosphatidylserine externalization, mitochondrial membrane potential loss, morphological changes, and DNA fragmentation. These effects support the conclusion that repurposed compounds and novel ruthenium-complexed chemotypes are potential candidates for antileishmanial drugs. This encourages the pursuit of further assays to maximize leishmanicidal effects while reducing cytotoxicity, aiming for a synergistic therapeutic approach.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/biology14121709/s1. Figure S1: Growth curve and assessment of MB concentrations on promastigote forms of L. amazonensis in vitro for 72 h; Figure S2: Flow cytometric evaluation of fluorescence on promastigote forms of L. amazonensis; Figure S3: Flow cytometric evaluation of cell death markers on promastigote forms of L. amazonensis; Figure S4: Cell death evaluation by flow cytometry on subpopulations of L. amazonensis promastigotes exposed to IC50 of the compounds for 24 h; Figure S5: Cell death evaluation by flow cytometry on promastigote forms of L. amazonensis exposed to 200 μM of MB for 24 h; Figure S6: Phosphatidylserine exposure in ex vivo amastigote forms of L. amazonensis; Figure S7: Cell death evaluation by flow cytometry on subpopulations of L. amazonensis ex vivo amastigote forms exposed to IC50 of the compounds for 24 h.

Author Contributions

Conceptualization, D.R.V.-d.-S., A.H.-P. and M.A.V.-S.; methodology, Z.N.d.R.; validation, D.R.V.-d.-S., J.A.-S., L.F.d.A.F. and N.V.-M.; formal analysis, D.R.V.-d.-S., J.A.-S., L.F.d.A.F., N.V.-M. and A.H.-P.; investigation, D.R.V.-d.-S., J.A-S., L.F.d.A.F. and N.V.-M.; resources, Z.N.d.R., M.d.N.C.S., A.H.-P., E.C.T.-S. and M.A.V.-S.; data curation, D.R.V.-d.-S. and M.A.V.-S.; writing—original draft preparation, D.R.V.-d.-S.; writing—review and editing, D.R.V.-d.-S., Z.N.d.R., M.d.N.C.S., A.H.-P., E.C.T.-S. and M.A.V.-S.; visualization, D.R.V.-d.-S., N.V.-M., E.C.T.-S. and M.A.V.-S.; supervision, E.C.T.-S. and M.A.V.-S.; project administration, M.A.V.-S.; funding acquisition, M.A.V.-S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was sponsored by the Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ; grant number 260003/001221/2023), the Programa Inova Fiocruz, and the Instituto Oswaldo Cruz (IOC/Fiocruz; IOC-008-FIO-22-2-51). Doctoral scholarships were awarded to D.R.V-D-S. (Conselho Nacional de Desenvolvimento Científico e Tecnológico—CNPq; 142350/2020-6) and N.V-M. (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior—CAPES). Postdoctorial scholarships were granted to J.A-S. (CNPq; 153198/2024-9) and L.F.A.F. (FAPERJ; E-26/200.400/2024). Senior researchers M.N.C.S. (CNPq; 312170/2023-8—CNE/FAPERJ; E26/203.969/2024), A.H-P. (CNPq; 310887/2023-2), E.C.T-S. (CNPq; 313188/2021-1—CNE/FAPERJ—E26/201.158/2022), and M.A.V.-S. (CNPq—314717/2020-0; CNE/FAPERJ—E26/204.392/2024) are productivity fellows of the CNPq and the Programa Cientista do Nosso Estado (CNE) of FAPERJ.

Institutional Review Board Statement

Genetic access was approved by SisGen (AD1C828). Animal studies were approved by CEUA—IOC/Fiocruz, Rio de Janeiro (L038-2017 A4)—in accordance with CONCEA guidelines.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

The authors thank C.M. Cascabulho and D.G. Beghini, technologists at the Flow Cytometry Facility—Unit of Multiparametric Analysis of Instituto Oswaldo Cruz (IOC/Fiocruz)—for their support, and M.B. Meuser, at the Laboratory of Cell and Molecular Biology (IOC/Fiocruz), for technical assistance with animal experiments.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zhu, Y.S.; Sun, Z.S.; Zheng, J.X.; Zhang, S.X.; Yin, J.X.; Zhao, H.Q.; Shen, H.M.; Baneth, G.; Chen, J.H.; Kassegne, K. Prevalence and Attributable Health Burdens of Vector-Borne Parasitic Infectious Diseases of Poverty, 1990–2021: Findings from the Global Burden of Disease Study 2021. Infect. Dis. Poverty 2024, 13, 96. [Google Scholar] [CrossRef] [PubMed]
  2. Valero, N.N.H.; Uriarte, M. Environmental and Socioeconomic Risk Factors Associated with Visceral and Cutaneous Leishmaniasis: A Systematic Review. Parasitol. Res. 2020, 119, 365–384. [Google Scholar] [CrossRef]
  3. Sasidharan, S.; Saudagar, P. Leishmaniasis: Where are We and Where are We Heading? Parasitol. Res. 2021, 120, 1541–1554. [Google Scholar] [CrossRef] [PubMed]
  4. WHO, World Health Organization. Leishmaniasis Overview. Available online: https://www.who.int/health-topics/leishmaniasis#tab=tab_1 (accessed on 8 June 2024).
  5. Silva, A.P.O.; Miranda, D.E.O.; Santos, M.A.B.; Guerra, N.R.; Marques, S.R.; Alves, L.C.; Ramos, R.A.N.; Carvalho, G.A. Phlebotomines in an Area Endemic for American Cutaneous Leishmaniasis in Northeastern Coast of Brazil. Rev. Bras. Parasitol. Vet. 2017, 26, 280–284. [Google Scholar] [CrossRef]
  6. Serafim, T.D.; Coutinho-Abreu, I.V.; Dey, R.; Kissinger, R.; Valenzuela, J.G.; Oliveira, F.; Kamhawi, S. Leishmaniasis: The Act of Transmission. Trends Parasitol. 2021, 37, 976–987. [Google Scholar] [CrossRef] [PubMed]
  7. Reimann, M.M.; Torres-Santos, E.C.; Souza, C.S.F.; Andrade-Neto, V.V.; Jansen, A.M.; Brazil, R.P.; Roque, A.L.R. Oral and Intragastric: New Routes of Infection by Leishmania braziliensis and Leishmania infantum? Pathogens 2022, 11, 688. [Google Scholar] [CrossRef] [PubMed]
  8. Daga, R.; Mishra, R.; Rohatgi, I. Leishmaniasis. Indian J. Crit. Care Med. 2021, 25, S166–S170. [Google Scholar] [CrossRef]
  9. WHO, World Health Organization. Leishmaniasis Key Facts. Available online: https://www.who.int/en/news-room/fact-sheets/detail/leishmaniasis (accessed on 8 June 2024).
  10. Barral, A.; Costa, J.M.L.; Bittencourt, A.L.; Barral-Netto, M.; Carvalho, E.M. Polar and Subpolar Diffuse Cutaneous Leishmaniasis in Brazil: Clinical and Immunopathologic Aspects. Int. J. Dermatol. 1995, 34, 474–479. [Google Scholar] [CrossRef] [PubMed]
  11. OPAS, Organización Panamericana de la Salud. Manual de Procedimientos para Vigilancia y Control de las Leishmaniasis en las Américas; Organización Panamericana de la Salud: Washington, DC, USA, 2019; ISBN 9789275320631. [Google Scholar]
  12. Pereira, V.; Barros, N.B.; Macedo, S.R.A.; Ferreira, A.S.; Kanis, L.A.; Nicolete, R. Drug-Containing Hydrophobic Dressings as a Topical Experimental Therapy for Cutaneous Leishmaniasis. J. Parasit. Dis. 2020, 44, 79–87. [Google Scholar] [CrossRef]
  13. Bennis, I.; De Brouwere, V.; Belrhiti, Z.; Sahibi, H.; Boelaert, M. Psychosocial Burden of Localised Cutaneous Leishmaniasis: A Scoping Review. BMC Public Health 2018, 18, 358. [Google Scholar] [CrossRef] [PubMed]
  14. Gómez, L.J.; Van Wijk, R.; Van Selm, L.; Rivera, A.; Barbosa, M.C.; Parisi, S.; Van Brakel, W.H.; Arevalo, J.; Quintero, W.; Waltz, M.; et al. Stigma, Participation Restriction and Mental Distress in Patients Affected by Leprosy, Cutaneous Leishmaniasis and Chagas Disease: A Pilot Study in Two Co-Endemic Regions of Eastern Colombia. Trans. R. Soc. Trop. Med. Hyg. 2019, 114, 476–482. [Google Scholar] [CrossRef] [PubMed]
  15. Volpedo, G.; Pacheco-Fernandez, T.; Holcomb, E.A.; Cipriano, N.; Cox, B.; Satoskar, A.R. Mechanisms of Immunopathogenesis in Cutaneous Leishmaniasis and Post Kala-Azar Dermal Leishmaniasis (PKDL). Front. Cell. Infect. Microbiol. 2021, 11, 685296. [Google Scholar] [CrossRef] [PubMed]
  16. Sundar, S.; Madhukar, P.; Kumar, R. Anti-Leishmanial Therapies: Overcoming Current Challenges with Emerging Therapies. Expert Rev. Anti-Infect. Ther. 2024, 7, 1–9. [Google Scholar] [CrossRef] [PubMed]
  17. Torres-Guerrero, E.; Quintanilla-Cedillo, M.R.; Ruiz-Esmenjaud, J.; Arenas, R. Leishmaniasis: A Review. F1000Research 2017, 6, 750. [Google Scholar] [CrossRef] [PubMed]
  18. Bastos, D.S.S.; Miranda, B.M.; Fialho Martins, T.V.; Guimarães Ervilha, L.O.; Souza, A.C.F.; Emerick, S.O.; Silva, A.C.; Novaes, R.D.; Neves, M.M.; Santos, E.C.; et al. Lipophosphoglycan-3 Recombinant Protein Vaccine Controls Hepatic Parasitism and Prevents Tissue Damage in Mice Infected by Leishmania infantum chagasi. Biomed. Pharmacother. 2020, 126, 110097. [Google Scholar] [CrossRef] [PubMed]
  19. Periferakis, A.; Caruntu, A.; Periferakis, A.-T.; Scheau, A.-E.; Badarau, I.A.; Caruntu, C.; Scheau, C. Availability, Toxicology and Medical Significance of Antimony. Int. J. Environ. Res. Public Health 2022, 19, 4669. [Google Scholar] [CrossRef] [PubMed]
  20. Oliveira, L.F.; Schubach, A.O.; Martins, M.M.; Passos, S.L.; Oliveira, R.V.; Marzochi, M.C.; Andrade, C.A. Systematic Review of the Adverse Effects of Cutaneous Leishmaniasis Treatment in the New World. Acta Trop. 2011, 118, 87–96. [Google Scholar] [CrossRef]
  21. Sundar, S.; Chakravarty, J. Antimony Toxicity. Int. J. Environ. Res. Public Health 2010, 7, 4267–4277. [Google Scholar] [CrossRef]
  22. Rijal, S.; Chappuis, F.; Singh, R.; Boelaert, M.; Loutan, L.; Koirala, S. Sodium Stibogluconate Cardiotoxicity and Safety of Generics. Trans. R. Soc. Trop. Med. Hyg. 2003, 97, 597–598. [Google Scholar] [CrossRef] [PubMed]
  23. Farina, J.M.; García-Martínez, C.E.; Saldarriaga, C.; Pérez, G.E.; Melo, M.B.; Wyss, F.; Sosa-Liprandi, Á.; Ortiz-Lopez, H.I.; Gupta, S.; López-Santi, R.; et al. Leishmaniasis y Corazón. Arch. Cardiol. Méx. 2022, 92, 85–93. [Google Scholar] [CrossRef] [PubMed]
  24. Kato, K.C.; Morais-Teixeira, E.; Reis, P.G.; Silva-Barcellos, N.M.; Salaün, P.; Campos, P.P.; Corrêa-Junior, J.D.; Rabello, A.; Demicheli, C.; Frézard, F. Hepatotoxicity of Pentavalent Antimonial Drug: Possible Role of Residual Sb(III) and Protective Effect of Ascorbic Acid. Antimicrob. Agents Chemother. 2014, 58, 481–488. [Google Scholar] [CrossRef]
  25. Lima, M.I.S.; Arruda, V.O.; Alves, E.V.C.; Azevedo, A.P.S.; Monteiro, S.G.; Pereira, S.R.F. Genotoxic Effects of the Antileishmanial Drug Glucantime®. Arch. Toxicol. 2010, 84, 227–232. [Google Scholar] [CrossRef] [PubMed]
  26. Khalil, E.A.G.; Ahmed, A.E.; Musa, A.E.; Hussein, M.M. Antimony-Induced Cerebellar Ataxia. Saudi Med. J. 2006, 27, 90–92. [Google Scholar] [PubMed]
  27. Maristany Bosch, M.; Cuervo, G.; Martín, E.M.; Heras, M.V.; Pérez, J.P.; Yélamos, S.M.; Fernández, N.S. Neurological Toxicity Due to Antimonial Treatment for Refractory Visceral Leishmaniasis. Clin. Neurophysiol. Pract. 2021, 6, 164–167. [Google Scholar] [CrossRef] [PubMed]
  28. Rodrigues, M.L.O.; Costa, R.S.; Souza, C.S.; Foss, N.T.; Roselino, A.M.F. Nephrotoxicity Attributed to Meglumine Antimoniate (Glucantime) in the Treatment of Generalized Cutaneous Leishmaniasis. Rev. Inst. Med. Trop. 1999, 41, 33–37. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, L.; Luo, C.; Zheng, D.; Wang, X.; Wang, R.; Ding, W.; Shen, Z.; Xue, P.; Yu, S.; Liu, Y.; et al. TRPML1 Contributes to Antimony-Induced Nephrotoxicity by Initiating Ferroptosis via Chaperone-Mediated Autophagy. Food Chem. Toxicol. 2024, 184, 114378. [Google Scholar] [CrossRef] [PubMed]
  30. Morgan, D.J.; Guimaraes, L.H.; Machado, P.R.L.; D’Oliveira, A.; Almeida, R.P.; Lago, E.L.; Faria, D.R.; Tafuri, W.L.; Dutra, W.O.; Carvalho, E.M. Cutaneous Leishmaniasis During Pregnancy: Exuberant Lesions and Potential Fetal Complications. Clin. Infect. Dis. 2007, 45, 478–482. [Google Scholar] [CrossRef]
  31. Marques, S.A.; Merlotto, M.R.; Ramos, P.M.; Marques, M.E.A. American Tegumentary Leishmaniasis: Severe Side Effects of Pentavalent Antimonial in a Patient with Chronic Renal Failure. An. Bras. Dermatol. 2019, 94, 355–357. [Google Scholar] [CrossRef]
  32. Brito, N.C.; Rabello, A.; Cota, G.F. Efficacy of Pentavalent Antimoniate Intralesional Infiltration Therapy for Cutaneous Leishmaniasis: A Systematic Review. PLoS ONE 2017, 12, e0184777. [Google Scholar] [CrossRef]
  33. Shmueli, M.; Ben-Shimol, S. Review of Leishmaniasis Treatment: Can We See the Forest through the Trees? Pharmacy 2024, 12, 30. [Google Scholar] [CrossRef]
  34. Roatt, B.M.; Cardoso, J.M.O.; Brito, R.C.F.; Coura-Vital, W.; Aguiar-Soares, R.D.O.; Reis, A.B. Recent Advances and New Strategies on Leishmaniasis Treatment. Appl. Microbiol. Biotechnol. 2020, 104, 8965–8977. [Google Scholar] [CrossRef]
  35. Ponte-Sucre, A.; Gamarro, F.; Dujardin, J.C.; Barrett, M.P.; López-Vélez, R.; García-Hernández, R.; Pountain, A.W.; Mwenechanya, R.; Papadopoulou, B. Drug Resistance and Treatment Failure in Leishmaniasis: A 21st Century Challenge. PLoS Negl. Trop. Dis. 2017, 11, e0006052. [Google Scholar] [CrossRef]
  36. Garza-Tovar, T.F.; Sacriste-Hernández, M.I.; Juárez-Durán, E.R.; Arenas, R. An Overview of the Treatment of Cutaneous Leishmaniasis. Fac. Rev. 2020, 9, 1–9. [Google Scholar] [CrossRef]
  37. DNDi, Drugs for Neglected Diseases Initiative. Leishmaniose Cutânea. Available online: https://dndial.org/doencas/leishmaniose-cutanea#dados/ (accessed on 8 June 2024).
  38. Ayyar, P.; Subramanian, U. Repurposing—Second Life for Drugs. Pharmacia 2022, 69, 51–59. [Google Scholar] [CrossRef]
  39. Sakate, R.; Kimura, T. Drug Repositioning Trends in Rare and Intractable Diseases. Drug Discov. Today 2022, 27, 1789–1795. [Google Scholar] [CrossRef] [PubMed]
  40. Buchholz, K.; Schirmer, R.H.; Eubel, J.K.; Akoachere, M.B.; Dandekar, T.; Becker, K.; Gromer, S. Interactions of Methylene Blue with Human Disulfide Reductases and Their Orthologues from Plasmodium falciparum. Antimicrob. Agents Chemother. 2008, 52, 183–191. [Google Scholar] [CrossRef] [PubMed]
  41. Appleby, B.; Cummings, J. Discovering New Treatments for Alzheimer’s Disease by Repurposing Approved Medications. Curr. Top. Med. Chem. 2013, 13, 2306–2327. [Google Scholar] [CrossRef] [PubMed]
  42. Jaeschke, H.; Akakpo, J.Y.; Umbaugh, D.S.; Ramachandran, A. Novel Therapeutic Approaches Against Acetaminophen-Induced Liver Injury and Acute Liver Failure. Toxicol. Sci. 2020, 174, 159–167. [Google Scholar] [CrossRef] [PubMed]
  43. Dabholkar, N.; Gorantla, S.; Dubey, S.K.; Alexander, A.; Taliyan, R.; Singhvi, G. Repurposing Methylene Blue in the Management of COVID-19: Mechanistic Aspects and Clinical Investigations. Biomed. Pharmacother. 2021, 142, 112023. [Google Scholar] [CrossRef] [PubMed]
  44. Maheshwari, A.; Seth, A.; Kaur, S.; Aneja, S.; Rath, B.; Basu, S.; Patel, R.; Dutta, A.K. Cumulative Cardiac Toxicity of Sodium Stibogluconate and Amphotericin B in Treatment of Kala-Azar. Pediatr. Infect. Dis. J. 2011, 30, 180–181. [Google Scholar] [CrossRef] [PubMed]
  45. Wiklund, L.; Basu, S.; Miclescu, A.; Wiklund, P.; Ronquist, G.; Sharma, H.S. Neuro- and Cardioprotective Effects of Blockade of Nitric Oxide Action by Administration of Methylene Blue. Ann. N. Y. Acad. Sci. 2007, 1122, 231–244. [Google Scholar] [CrossRef]
  46. Llanos-Cuentas, A.; Valencia, B.M.; Petersen, C.A. Neurological Manifestations of Human Leishmaniasis. In Handbook of Clinical Neurology; Elsevier B.V.: Amsterdam, The Netherlands, 2013; Volume 114, pp. 193–198. [Google Scholar]
  47. Maia, C.S.F.; Monteiro, M.C.; Gavioli, E.C.; Oliveira, F.R.; Oliveira, G.B.; Romão, P.R.T. Neurological Disease in Human and Canine Leishmaniasis—Clinical Features and Immunopathogenesis. Parasite Immunol. 2015, 37, 385–393. [Google Scholar] [CrossRef] [PubMed]
  48. Giannuzzi, A.P.; Ricciardi, M.; De Simone, A.; Gernone, F. Neurological Manifestations in Dogs Naturally Infected by Leishmania infantum: Descriptions of 10 Cases and a Review of the Literature. J. Small Anim. Pract. 2017, 58, 125–138. [Google Scholar] [CrossRef]
  49. Petersen, C.A.; Greenlee, M.H.W. Neurologic Manifestations of Leishmania spp. Infection. J. Neuroparasitol. 2011, 2, 1–9. [Google Scholar] [CrossRef]
  50. Gureev, A.P.; Sadovnikova, I.S.; Popov, V.N. Molecular Mechanisms of the Neuroprotective Effect of Methylene Blue. Biochemistry 2022, 87, 940–956. [Google Scholar] [CrossRef]
  51. Lin, Z.H.; Wang, S.Y.; Chen, L.L.; Zhuang, J.Y.; Ke, Q.F.; Xiao, D.R.; Lin, W.P. Methylene Blue Mitigates Acute Neuroinflammation after Spinal Cord Injury through Inhibiting NLRP3 Inflammasome Activation in Microglia. Front. Cell. Neurosci. 2017, 11, 391. [Google Scholar] [CrossRef] [PubMed]
  52. Pinto, J.G.; Martins, J.F.S.; Pereira, A.H.C.; Mittmann, J.; Raniero, L.J.; Ferreira-Strixino, J. Evaluation of Methylene Blue as Photosensitizer in Promastigotes of Leishmania major and Leishmania braziliensis. Photodiagn. Photodyn. Ther. 2017, 18, 325–330. [Google Scholar] [CrossRef] [PubMed]
  53. Fall, B.; Madamet, M.; Diawara, S.; Briolant, S.; Wade, K.A.; Lo, G.; Nakoulima, A.; Fall, M.; Bercion, R.; Kounta, M.B.; et al. Ex vivo Activity of Proveblue, a Methylene Blue, against Field Isolates of Plasmodium falciparum in Dakar, Senegal from 2013–2015. Int. J. Antimicrob. Agents 2017, 50, 155–158. [Google Scholar] [CrossRef]
  54. Fagundes, J.; Sakane, K.K.; Bhattacharjee, T.; Pinto, J.G.; Ferreira, I.; Raniero, L.J.; Ferreira-Strixino, J. Evaluation of Photodynamic Therapy with Methylene Blue, by the Fourier Transform Infrared Spectroscopy (FT-IR) in Leishmania major—In vitro. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2019, 207, 229–235. [Google Scholar] [CrossRef]
  55. Sakane, K.K.; Bhattacharjee, T.; Fagundes, J.; Marcolino, L.M.C.; Ferreira, I.; Pinto, J.G.; Ferreira-Strixino, J. Biochemical Changes in Leishmania braziliensis after Photodynamic Therapy with Methylene Blue Assessed by the Fourier Transform Infrared Spectroscopy. Lasers Med. Sci. 2020, 36, 821–827. [Google Scholar] [CrossRef]
  56. Ozlem-Caliskan, S.; Ertabaklar, H.; Bilgin, M.D.; Ertug, S. Evaluation of Photodynamic Therapy against Leishmania tropica Promastigotes Using Different Photosensitizers. Photodermatol. Photoimmunol. Photomed. 2022, 38, 354–364. [Google Scholar] [CrossRef] [PubMed]
  57. Aureliano, D.P.; Lindoso, J.A.L.; Soares, S.R.C.; Takakura, C.F.H.; Pereira, T.M.; Ribeiro, M.S. Cell Death Mechanisms in Leishmania amazonensis Triggered by Methylene Blue-Mediated Antiparasitic Photodynamic Therapy. Photodiagn. Photodyn. Ther. 2018, 23, 1–8. [Google Scholar] [CrossRef]
  58. Cabral, F.V.; Sabino, C.P.; Dimmer, J.A.; Sauter, I.P.; Cortez, M.J.; Ribeiro, M.S. Preclinical Investigation of Methylene Blue-mediated Antimicrobial Photodynamic Therapy on Leishmania Parasites Using Real-Time Bioluminescence. Photochem. Photobiol. 2020, 96, 604–610. [Google Scholar] [CrossRef]
  59. Peloi, L.S.; Biondo, C.E.G.; Kimura, E.; Politi, M.J.; Lonardoni, M.V.C.; Aristides, S.M.A.; Dorea, R.C.C.; Hioka, N.; Silveira, T.G.V. Photodynamic Therapy for American Cutaneous Leishmaniasis: The Efficacy of Methylene Blue in Hamsters Experimentally Infected with Leishmania (Leishmania) amazonensis. Exp. Parasitol. 2011, 128, 353–356. [Google Scholar] [CrossRef]
  60. Song, D.; Lindoso, J.A.L.; Oyafuso, L.K.; Kanashiro, E.H.Y.; Cardoso, J.L.; Uchoa, A.F.; Tardivo, J.P.; Baptista, M.S. Photodynamic Therapy Using Methylene Blue to Treat Cutaneous Leishmaniasis. Photomed. Laser Surg. 2011, 29, 711–715. [Google Scholar] [CrossRef] [PubMed]
  61. Alamon-Reig, F.; Martí-Martí, I.; Loughlin, C.R.M.; Garcia, A.; Carrera, C.; Aguilera-Peiró, P. Successful Treatment of Facial Cutaneous Leishmaniasis with Photodynamic Therapy. Indian J. Dermatol. Venereol. Leprol. 2022, 88, 667. [Google Scholar] [CrossRef] [PubMed]
  62. Iniguez, E.; Varela-Ramirez, A.; Martínez, A.; Torres, C.L.; Sánchez-Delgado, R.A.; Maldonado, R.A. Ruthenium-Clotrimazole Complex has Significant Efficacy in the Murine Model of Cutaneous Leishmaniasis. Acta Trop. 2016, 164, 402–410. [Google Scholar] [CrossRef] [PubMed]
  63. Costa, M.S.; Gonçalves, Y.G.; Nunes, D.C.O.; Napolitano, D.R.; Maia, P.I.S.; Rodrigues, R.S.; Rodrigues, V.M.; Von Poelhsitz, G.; Yoneyama, K.A.G. Anti-Leishmania Activity of New Ruthenium(II) Complexes: Effect on Parasite-Host Interaction. J. Inorg. Biochem. 2017, 175, 225–231. [Google Scholar] [CrossRef] [PubMed]
  64. Miranda, V.M.; Costa, M.S.; Guilardi, S.; Machado, A.E.H.; Ellena, J.A.; Tudini, K.A.G.; Von Poelhsitz, G. In vitro Leishmanicidal Activity and Theoretical Insights into Biological Action of Ruthenium(II) Organometallic Complexes Containing Anti-Inflammatories. BioMetals 2018, 31, 1003–1017. [Google Scholar] [CrossRef] [PubMed]
  65. Nascimento, N.R.F.; Aguiar, F.L.N.; Santos, C.F.; Costa, A.M.L.; Hardoim, D.J.; Calabrese, K.S.; Almeida-Souza, F.; Sousa, E.H.S.; Lopes, L.G.F.; Teixeira, M.J.; et al. In vitro and in vivo Leishmanicidal Activity of a Ruthenium Nitrosyl Complex against Leishmania (Viannia) braziliensis. Acta Trop. 2019, 192, 61–65. [Google Scholar] [CrossRef] [PubMed]
  66. Valigurová, A.; Kolářová, I. Unrevealing the Mystery of Latent Leishmaniasis: What Cells Can Host Leishmania? Pathogens 2023, 12, 246. [Google Scholar] [CrossRef] [PubMed]
  67. Gomes-Junior, R.A.; Silva, R.S.; Lima, R.G.; Vannier-Santos, M.A. Antifungal Mechanism of [RuIII(NH3)4catechol]+ Complex on Fluconazole-Resistant Candida tropicalis. FEMS Microbiol. Lett. 2017, 364, fnx073. [Google Scholar] [CrossRef] [PubMed]
  68. Leon, L.; Torres-Santos, E.C. Differents Aspects on Chemotherapy of Trypanosomatids; Nova Science Pub Inc.: Hauppauge, NY, USA, 2017; ISBN 1536108502. [Google Scholar]
  69. Santos, C.C.; Zhang, H.; Batista, M.M.; Oliveira, G.M.; Demarque, K.C.; Silva, N.L.; Moreira, O.C.; Ogungbe, I.V.; Soeiro, M.D.N.C. Phenotypic Investigation of 4-Nitrophenylacetyl- and 4-Nitro-1H-Imidazoyl-Based Compounds as Antileishmanial Agents. Parasitology 2022, 149, 490–495. [Google Scholar] [CrossRef] [PubMed]
  70. Araujo-Lima, C.F.; Christoni, L.S.A.; Justo, G.; Soeiro, M.N.C.; Aiub, C.A.F.; Felzenszwalb, I. Atorvastatin Downregulates In vitro Methyl Methanesulfonate and Cyclophosphamide Alkylation-Mediated Cellular and DNA Injuries. Oxid. Med. Cell. Longev. 2018, 2018, 7820890. [Google Scholar] [CrossRef] [PubMed]
  71. Rottini, M.M.; Amaral, A.C.F.; Ferreira, J.L.P.; Oliveira, E.S.C.; Silva, J.R.A.; Taniwaki, N.N.; Santos, A.R.; Almeida-Souza, F.; Souza, C.S.F.; Calabrese, K.S. Endlicheria bracteolata (Meisn.) Essential Oil as a Weapon Against Leishmania amazonensis: In vitro Assay. Molecules 2019, 24, 2525. [Google Scholar] [CrossRef]
  72. Romanha, A.J.; Castro, S.L.; Soeiro, M.N.C.; Lannes-Vieira, J.; Ribeiro, I.; Talvani, A.; Bourdin, B.; Blum, B.; Olivieri, B.; Zani, C.; et al. In vitro and in vivo Experimental Models for Drug Screening and Development for Chagas Disease. Mem. Inst. Oswaldo Cruz 2010, 105, 233–238. [Google Scholar] [CrossRef]
  73. Cardoso-Santos, C.; Meuser Batista, M.; Inam Ullah, A.; Rama Krishna Reddy, T.; Soeiro, M.D.N.C. Drug Screening Using Shape-Based Virtual Screening and in vitro Experimental Models of Cutaneous Leishmaniasis. Parasitology 2021, 148, 98–104. [Google Scholar] [CrossRef] [PubMed]
  74. Kumar, P.; Nagarajan, A.; Uchil, P.D. Analysis of Cell Viability by the AlamarBlue Assay. Cold Spring Harb. Protoc. 2018, 2018, pdb.prot095489. [Google Scholar] [CrossRef] [PubMed]
  75. Balanco, J.M.F.; Costa Moreira, M.E.; Bonomo, A.; Bozza, P.T.; Amarante-Mendes, G.; Pirmez, C.; Barcinski, M.A. Apoptotic Mimicry by an Obligate Intracellular Parasite Downregulates Macrophage Microbicidal Activity. Curr. Biol. 2001, 11, 1870–1873. [Google Scholar] [CrossRef]
  76. Wanderley, J.L.M.; Damatta, R.A.; Barcinski, M.A. Apoptotic Mimicry as a Strategy for the Establishment of Parasitic Infections: Parasite- and Host-Derived Phosphatidylserine as Key Molecule. Cell Commun. Signal. 2020, 18, 10. [Google Scholar] [CrossRef]
  77. Wanderley, J.L.M.; Silva, L.H.P.; Deolindo, P.; Soong, L.; Borges, V.M.; Prates, D.B.; Souza, A.P.A.; Barral, A.; Balanco, J.M.F.; Nascimento, M.T.C.; et al. Cooperation between Apoptotic and Viable Metacyclics Enhances the Pathogenesis of Leishmaniasis. PLoS ONE 2009, 4, e5733. [Google Scholar] [CrossRef]
  78. Weingärtner, A.; Kemmer, G.; Müller, F.D.; Zampieri, R.A.; Santos, M.G.; Schiller, J.; Pomorski, T.G. Leishmania Promastigotes Lack Phosphatidylserine but Bind Annexin V upon Permeabilization or Miltefosine Treatment. PLoS ONE 2012, 7, e42070. [Google Scholar] [CrossRef] [PubMed]
  79. Ridley, M. The Power of Parasites. In The Red Queen: Sex and the Evolution of Human Nature; Viking Books: London, UK, 1993; p. 310. ISBN 0-06-055657-9. [Google Scholar]
  80. Neuenschwander, A.; Rocha, V.P.C.; Bastos, T.M.; Marcourt, L.; Morin, H.; Rocha, C.Q.; Grimaldi, G.B.; Sousa, K.A.F.; Borges, J.N.; Rivara-Minten, E.; et al. Production of Highly Active Antiparasitic Compounds from the Controlled Halogenation of the Arrabidaea brachypoda Crude Plant Extract. J. Nat. Prod. 2020, 83, 2631–2640. [Google Scholar] [CrossRef] [PubMed]
  81. Carneiro, L.S.A.; Almeida-Souza, F.; Lopes, Y.S.C.; Novas, R.C.V.; Santos, K.B.A.; Ligiero, C.B.P.; Calabrese, K.S.; Buarque, C.D. Synthesis of 3-Aryl-4-(N-Aryl)Aminocoumarins via Photoredox Arylation and the Evaluation of their Biological Activity. Bioorg. Chem. 2021, 114, 105141. [Google Scholar] [CrossRef] [PubMed]
  82. Bortoleti, B.T.S.; Gonçalves, M.D.; Tomiotto-Pellissier, F.; Contato, V.M.; Silva, T.F.; Matos, R.L.N.; Detoni, M.B.; Rodrigues, A.C.J.; Carloto, A.C.; Lazarin, D.B.; et al. Solidagenone Acts on Promastigotes of L. amazonensis by Inducing Apoptosis-like Processes on Intracellular Amastigotes by IL-12p70/ROS/NO Pathway Activation. Phytomedicine 2021, 85, 153536. [Google Scholar] [CrossRef] [PubMed]
  83. Silva-Silva, J.V.; Moreira, R.F.; Watanabe, L.A.; Souza, C.S.F.; Hardoim, D.J.; Taniwaki, N.N.; Bertho, A.L.; Teixeira, K.F.; Cenci, A.R.; Doring, T.H.; et al. Monomethylsulochrin Isolated from Biomass Extract of Aspergillus sp. against Leishmania amazonensis: In vitro Biological Evaluation and Molecular Docking. Front. Cell. Infect. Microbiol. 2022, 12, 974910. [Google Scholar] [CrossRef] [PubMed]
  84. Silva-Silva, J.V.; Moragas-Tellis, C.J.; Chagas, M.S.S.; Souza, P.V.R.; Moreira, D.L.; Hardoim, D.J.; Taniwaki, N.N.; Costa, V.F.A.; Bertho, A.L.; Brondani, D.; et al. Carajurin Induces Apoptosis in Leishmania amazonensis Promastigotes through Reactive Oxygen Species Production and Mitochondrial Dysfunction. Pharmaceuticals 2022, 15, 331. [Google Scholar] [CrossRef] [PubMed]
  85. Monteiro, J.; Passero, L.F.D.; Jesus, J.A.; Laurenti, M.D.; Lago, J.H.G.; Soares, M.G.; Batista, A.N.L.; Batista, J.M.; Sartorelli, P. Absolute Configuration and Antileishmanial Activity of (–)-Cyclocolorenone Isolated from Duguetia lanceolata (Annonaceae). Curr. Top. Med. Chem. 2022, 22, 1626–1633. [Google Scholar] [CrossRef] [PubMed]
  86. Paes, S.S.; Silva-Silva, J.V.; Gomes, P.W.P.; Silva, L.O.; Costa, A.P.L.; Lopes Júnior, M.L.; Hardoim, D.J.; Moragas-Tellis, C.J.; Taniwaki, N.N.; Bertho, A.L.; et al. (-)-5-Demethoxygrandisin B a New Lignan from Virola Surinamensis (Rol.) Warb. Leaves: Evaluation of the Leishmanicidal Activity by in vitro and in silico Approaches. Pharmaceutics 2023, 15, 2292. [Google Scholar] [CrossRef] [PubMed]
  87. Santiago-Silva, K.M.; Bortoleti, B.T.S.; Oliveira, L.N.; Maia, F.L.A.; Castro, J.C.; Costa, I.C.; Lazarin, D.B.; Wardell, J.L.; Wardell, S.M.S.V.; Albuquerque, M.G.; et al. Antileishmanial Activity of 4,8-Dimethoxynaphthalenyl Chalcones on Leishmania amazonensis. Antibiotics 2022, 11, 1402. [Google Scholar] [CrossRef]
  88. Santiago-Silva, K.M.; Bortoleti, B.T.S.; Brito, T.O.; Costa, I.C.; Lima, C.H.S.; Macedo, F.; Miranda-Sapla, M.M.; Pavanelli, W.R.; Bispo, M.L.F. Exploring the Antileishmanial Activity of N1, N2-Disubstituted-Benzoylguanidines: Synthesis and Molecular Modeling Studies. J. Biomol. Struct. Dyn. 2022, 40, 11495–11510. [Google Scholar] [CrossRef]
  89. Camargo, P.G.; Bortoleti, B.T.S.; Fabris, M.; Gonçalves, M.D.; Tomiotto-Pellissier, F.; Costa, I.N.; Conchon-Costa, I.; Lima, C.H.S.; Pavanelli, W.R.; Bispo, M.L.F.; et al. Thiohydantoins as Anti-Leishmanial Agents: In vitro Biological Evaluation and Multi-Target Investigation by Molecular Docking Studies. J. Biomol. Struct. Dyn. 2022, 40, 3213–3222. [Google Scholar] [CrossRef]
  90. Oliveira, S.S.C.; Santos, V.S.; Devereux, M.; McCann, M.; Santos, A.L.S.; Branquinha, M.H. The Anti-Leishmania amazonensis and Anti-Leishmania chagasi Action of Copper(II) and Silver(I) 1,10-Phenanthroline-5,6-Dione Coordination Compounds. Pathogens 2023, 12, 70. [Google Scholar] [CrossRef] [PubMed]
  91. Maquiaveli, C.C.; Silva, E.R.; Jesus, B.H.; Monteiro, C.E.O.; Navarro, T.R.; Branco, L.O.P.; Santos, I.S.; Reis, N.F.; Portugal, A.B.; Wanderley, J.L.M.; et al. Design and Synthesis of New Anthranyl Phenylhydrazides: Antileishmanial Activity and Structure–Activity Relationship. Pharmaceuticals 2023, 16, 1120. [Google Scholar] [CrossRef] [PubMed]
  92. Gomes-Alves, A.G.; Duarte, M.; Cruz, T.; Castro, H.; Lopes, F.; Moreira, R.; Ressurreição, A.S.; Tomás, A.M. Biological Evaluation and Mechanistic Studies of Quinolin-(1H)-Imines as a New Chemotype against Leishmaniasis. Antimicrob. Agents Chemother. 2021, 65, e01513-20. [Google Scholar] [CrossRef] [PubMed]
  93. Present, C.; Girão, R.D.; Lin, C.; Caljon, G.; Van Calenbergh, S.; Moreira, O.; Ruivo, L.A.S.; Batista, M.M.; Azevedo, R.; Batista, D.G.J.; et al. N6-Methyltubercidin Gives Sterile Cure in a Cutaneous Leishmania amazonensis Mouse Model. Parasitology 2024, 151, 506–513. [Google Scholar] [CrossRef] [PubMed]
  94. Roquero, I.; Cantizani, J.; Cotillo, I.; Manzano, M.P.; Kessler, A.; Martín, J.J.; McNamara, C.W. Novel Chemical Starting Points for Drug Discovery in Leishmaniasis and Chagas Disease. Int. J. Parasitol. Drugs Drug Resist. 2019, 10, 58–68. [Google Scholar] [CrossRef]
  95. Ferreira, R.A.A.; Junior, C.O.R.; Martinez, P.D.G.; Koovits, P.J.; Soares, B.M.; Ferreira, L.L.G.; Michelan-Duarte, S.; Chelucci, R.C.; Andricopulo, A.D.; Galuppo, M.K.; et al. 2-Aminobenzimidazoles for Leishmaniasis: From Initial Hit Discovery to in vivo Profiling. PLoS Negl. Trop. Dis. 2021, 15, e0009196. [Google Scholar] [CrossRef] [PubMed]
  96. Katsuno, K.; Burrows, J.N.; Duncan, K.; Van Huijsduijnen, R.H.; Kaneko, T.; Kita, K.; Mowbray, C.E.; Schmatz, D.; Warner, P.; Slingsby, B.T. Hit and Lead Criteria in Drug Discovery for Infectious Diseases of the Developing World. Nat. Rev. Drug Discov. 2015, 14, 751–758. [Google Scholar] [CrossRef]
  97. Liu, D.; Uzonna, J.E. The Early Interaction of Leishmania with Macrophages and Dendritic Cells and Its Influence on the Host Immune Response. Front. Cell. Infect. Microbiol. 2012, 2, 83. [Google Scholar] [CrossRef] [PubMed]
  98. Duque, G.A.; Descoteaux, A. Macrophage Cytokines: Involvement in Immunity and Infectious Diseases. Front. Immunol. 2014, 5, 491. [Google Scholar] [CrossRef]
  99. Menezes, J.P.; Saraiva, E.M.; Rocha-Azevedo, B. The Site of the Bite: Leishmania Interaction with Macrophages, Neutrophils and the Extracellular Matrix in the Dermis. Parasites Vectors 2016, 9, 264. [Google Scholar] [CrossRef] [PubMed]
  100. Atri, C.; Guerfali, F.Z.; Laouini, D. Role of Human Macrophage Polarization in Inflammation during Infectious Diseases. Int. J. Mol. Sci. 2018, 19, 1801. [Google Scholar] [CrossRef] [PubMed]
  101. Davis, A. Comparative Trials of Antimonial Drugs in Urinary Schistosomiasis. Bull. World Health Organ. 1968, 38, 197–227. [Google Scholar] [PubMed]
  102. Saad, K.A.; Abdalla, I.O.; Alkailani, H.A.; Elbakush, A.M.; Zreiba, Z.A. Evaluation of Hepatotoxicity Effect of Sodium Stibogluconate (Pentostam) in Mice Model. Al-Mukhtar J. Sci. 2022, 37, 22–28. [Google Scholar] [CrossRef]
  103. Oliveira, A.L.L.; Brustoloni, Y.M.; Fernandes, T.D.; Dorval, M.E.C.; Cunha, R.V.; Bóia, M.N. Severe Adverse Reactions to Meglumine Antimoniate in the Treatment of Visceral Leishmaniasis: A Report of 13 Cases in the Southwestern Region of Brazil. Trop. Doct. 2009, 39, 180–182. [Google Scholar] [CrossRef]
  104. Rakhshan, A.; Kamel, B.R.; Saffaei, A.; Tavakoli-Ardekani, M. Hepatotoxicity Induced by Azole Antifungal Agents: A Review Study. Iran. J. Pharm. Res. 2023, 22, e130336. [Google Scholar] [CrossRef] [PubMed]
  105. Abajo, F.J.; Carcas, A.J. Fixed Drug Eruption Associated with DM Iadce Pa Anonsm. Br. Med. J. (Clin. Res. Ed.) 1986, 293, 1986. [Google Scholar]
  106. Fanos, V.; Cataldi, L. Amphotericin B-Induced Nephrotoxicity: A Review. J. Chemother. 2000, 12, 463–470. [Google Scholar] [CrossRef]
  107. Deray, G.; Mercadal, L.; Bagnis, C. Amphotericin B Nephrotoxicity. Nephrologie 2002, 23, 119–122. [Google Scholar] [CrossRef] [PubMed]
  108. Patel, G.P.; Crank, C.W.; Leikin, J.B. An Evaluation of Hepatotoxicity and Nephrotoxicity of Liposomal Amphotericin B (L-AMB). J. Med. Toxicol. 2011, 7, 12–15. [Google Scholar] [CrossRef] [PubMed]
  109. Pradhan, S.; Schwartz, R.A.; Patil, A.; Grabbe, S.; Goldust, M. Treatment Options for Leishmaniasis. Clin. Exp. Dermatol. 2022, 47, 516–521. [Google Scholar] [CrossRef] [PubMed]
  110. Mcgwire, B.S.; Satoskar, A.R. Leishmaniasis: Clinical Syndromes and Treatment. QJM Int. J. Med. 2014, 107, 7–14. [Google Scholar] [CrossRef] [PubMed]
  111. Prabhavalkar, S.; Masengu, A.; O’Rourke, D.; Shields, J.; Courtney, A. Nebulized Pentamidine-Induced Acute Renal Allograft Dysfunction. Case Rep. Transplant. 2013, 2013, 907593. [Google Scholar] [CrossRef]
  112. Matos, A.P.S.; Viçosa, A.L.; Ré, M.I.; Ricci-Júnior, E.; Holandino, C. A Review of Current Treatments Strategies Based on Paromomycin for Leishmaniasis. J. Drug Deliv. Sci. Technol. 2020, 57, 101664. [Google Scholar] [CrossRef]
  113. Sundar, S.; Jha, T.K.; Thakur, C.P.; Engel, J.; Sindermann, H.; Fischer, C.; Junge, K.; Bryceson, A.; Berman, J. Oral Miltefosine for Indian Visceral Leishmaniasis. N. Engl. J. Med. 2002, 347, 1739–1746. [Google Scholar] [CrossRef]
  114. Olliaro, P.L.; Guerin, P.J.; Gerstl, S.; Haaskjold, A.A.; Rottingen, J.A.; Sundar, S. Treatment Options for Visceral Leishmaniasis: A Systematic Review of Clinical Studies Done in India, 1980–2004. Lancet Infect. Dis. 2005, 5, 763–774. [Google Scholar] [CrossRef]
  115. Minodier, P.; Jurquet, A.L.; Noël, G.; Uters, M.; Laporte, R.; Garnier, J.M. Le Traitement Des Leishmanioses. Arch. Pediatr. 2010, 17, 838–839. [Google Scholar] [CrossRef] [PubMed]
  116. Chakravarty, J.; Sundar, S. Current and Emerging Medications for the Treatment of Leishmaniasis. Expert Opin. Pharmacother. 2019, 20, 1251–1265. [Google Scholar] [CrossRef] [PubMed]
  117. Castell, J.V.; Jover, R.; Martínez-Jiménez, C.P.; Gómez-Lechón, M.J. Hepatocyte Cell Lines: Their Use, Scope and Limitations in Drug Metabolism Studies. Expert Opin. Drug Metab. Toxicol. 2006, 2, 183–212. [Google Scholar] [CrossRef]
  118. Arzumanian, V.A.; Kiseleva, O.I.; Poverennaya, E.V. The Curious Case of the HepG2 Cell Line: 40 Years of Expertise. Int. J. Mol. Sci. 2021, 22, 13135. [Google Scholar] [CrossRef]
  119. Hameed, H.; King, E.F.B.; Doleckova, K.; Bartholomew, B.; Hollinshead, J.; Mbye, H.; Ullah, I.; Walker, K.; Van Veelen, M.; Abou-Akkada, S.S.; et al. Temperate Zone Plant Natural Products—A Novel Resource for Activity against Tropical Parasitic Diseases. Pharmaceuticals 2021, 14, 227. [Google Scholar] [CrossRef]
  120. Araújo, I.A.C.; De Paula, R.C.; Alves, C.L.; Faria, K.F.; Oliveira, M.M.; Takarada, G.G.M.; Dias, E.M.F.A.; Oliveira, A.B.; Silva, S.M. In Vitro Efficacy of Isoflavonoids and Terpenes against Leishmania (Leishmania) infantum and L. amazonensis. Exp. Parasitol. 2022, 242, 108383. [Google Scholar] [CrossRef]
  121. Gabelova, A.; Kozics, K.; Kapka-Skrzypczak, L.; Kruszewski, M.; Sramkova, M. Nephrotoxicity: Topical Issue. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2019, 845, 402988. [Google Scholar] [CrossRef] [PubMed]
  122. Pluta, K.; Jeleń, M.; Morak-Młodawska, B.; Zimecki, M.; Artym, J.; Kocięba, M.; Zaczyńska, E. Azaphenothiazines—Promising Phenothiazine Derivatives. An Insight into Nomenclature, Synthesis, Structure Elucidation and Biological Properties. Eur. J. Med. Chem. 2017, 138, 774–806. [Google Scholar] [CrossRef] [PubMed]
  123. Cavalcante-Costa, V.S.; Costa-Reginaldo, M.; Queiroz-Oliveira, T.; Oliveira, A.C.S.; Couto, N.F.; Dos Anjos, D.O.; Lima-Santos, J.; Andrade, L.O.; Horta, M.F.; Castro-Gomes, T. Leishmania amazonensis Hijacks Host Cell Lysosomes Involved in Plasma Membrane Repair to Induce Invasion in Fibroblasts. J. Cell Sci. 2019, 132, jcs226183. [Google Scholar] [CrossRef]
  124. Yektaeian, N.; Zare, S.; Radfar, A.H.; Hatam, G. Superparamagnetic Iron Oxide-Labeled Leishmania major can be Traced in Fibroblasts. J. Parasitol. Res. 2023, 2023, 7628912. [Google Scholar] [CrossRef] [PubMed]
  125. García-Davis, S.; López-Arencibia, A.; Bethencourt-Estrella, C.J.; Nicolás-Hernández, D.S.; Viveros-Valdez, E.; Díaz-Marrero, A.R.; Fernández, J.J.; Lorenzo-Morales, J.; Piñero, J.E. Laurequinone, a Lead Compound against Leishmania. Mar. Drugs 2023, 21, 333. [Google Scholar] [CrossRef]
  126. Parise-Filho, R.; Pasqualoto, K.F.M.; Magri, F.M.M.; Ferreira, A.K.; Silva, B.A.V.G.; Damião, M.C.F.C.B.; Tavares, M.T.; Azevedo, R.A.; Auada, A.V.V.; Polli, M.C.; et al. Dillapiole as Antileishmanial Agent: Discovery, Cytotoxic Activity and Preliminary SAR Studies of Dillapiole Analogues. Arch. Pharm. 2012, 345, 934–944. [Google Scholar] [CrossRef] [PubMed]
  127. Sá, G.C.S.; Silva, L.B.; Bezerra, P.V.V.; Silva, M.A.F.; Inacio, C.L.S.; Paiva, W.S.; Macedo e Silva, V.P.; Cordeiro, L.V.; Oliveira, J.W.F.; Silva, M.S.; et al. Tephrosia toxicaria (Sw.) Pers. Extracts: Screening by Examining Aedicidal Action under Laboratory and Field Conditions along with Its Antioxidant, Antileishmanial, and Antimicrobial Activities. PLoS ONE 2023, 18, e0275835. [Google Scholar] [CrossRef]
  128. Fermiano, M.H.; Neves, A.R.; Silva, F.; Barros, M.S.A.; Vieira, C.B.; Stein, A.L.; Frizon, T.E.A.; Braga, A.L.; Arruda, C.C.P.; Parisotto, E.B.; et al. Selenium-Containing (Hetero)Aryl Hybrids as Potential Antileishmanial Drug Candidates: In vitro Screening against L. amazonensis. Biomedicines 2024, 12, 213. [Google Scholar] [CrossRef] [PubMed]
  129. Braga, S.F.P.; Alves, É.V.P.; Ferreira, R.S.; Fradico, J.R.B.; Lage, P.S.; Duarte, M.C.; Ribeiro, T.G.; Júnior, P.A.S.; Romanha, A.J.; Tonini, M.L.; et al. Synthesis and Evaluation of the Antiparasitic Activity of Bis-(Arylmethylidene) Cycloalkanones. Eur. J. Med. Chem. 2014, 71, 282–289. [Google Scholar] [CrossRef] [PubMed]
  130. Carvalho, S.G.; Siqueira, L.A.; Zanini, M.S.; Matos, A.P.S.; Quaresma, C.H.; Silva, L.M.; Andrade, S.F.; Severi, J.A.; Villanova, J.C.O. Physicochemical and in vitro Biological Evaluations of Furazolidone-Based β-Cyclodextrin Complexes in Leishmania amazonensis. Res. Vet. Sci. 2018, 119, 143–153. [Google Scholar] [CrossRef]
  131. Costa, A.M.B.; Silva, A.R.S.T.; Santos, A.J.; Galvão, J.G.; Andrade-Neto, V.V.; Torres-Santos, E.C.; Ueki, M.M.; Almeida, L.E.; Sarmento, V.H.V.; Dolabella, S.S.; et al. Thermosensitive System Formed by Poloxamers Containing Carvacrol: An Effective Carrier System against Leishmania amazonensis. Acta Trop. 2023, 237, 106744. [Google Scholar] [CrossRef] [PubMed]
  132. Cabral, F.V.; Riahi, M.; Persheyev, S.; Lian, C.; Cortez, M.; Samuel, I.D.W.; Ribeiro, M.S. Photodynamic Therapy Offers a Novel Approach to Managing Miltefosine-Resistant Cutaneous Leishmaniasis. Biomed. Pharmacother. 2024, 177, 116881. [Google Scholar] [CrossRef] [PubMed]
  133. Sbeghen, M.R.; Voltarelli, E.M.; Campois, T.G.; Kimura, E.; Aristides, S.M.A.; Hernandes, L.; Caetano, W.; Hioka, N.; Lonardoni, M.V.C.; Silveira, T.G.V. Topical and Intradermal Efficacy of Photodynamic Therapy with Methylene Blue and Light-Emitting Diode in the Treatment of Cutaneous Leishmaniasis Caused by Leishmania braziliensis. J. Lasers Med. Sci. 2015, 6, 106–111. [Google Scholar] [CrossRef]
  134. Oliveira, S.; Trahamane, E.J.O.; Monteiro, J.; Santos, G.P.; Crugeira, P.; Sampaio, F.; Oliveira, C.; Neto, M.B.; Pinheiro, A. Leishmanicidal Effect of Antiparasitic Photodynamic Therapy—ApPDT on Infected Macrophages. Lasers Med. Sci. 2017, 32, 1959–1964. [Google Scholar] [CrossRef] [PubMed]
  135. Volpe, R.A.F.N.; Navasconi, T.R.; Reis, V.N.; Hioka, N.; Becker, T.C.A.; Lonardoni, M.V.C.; Aristides, S.M.A.; Silveira, T.G.V. Photodynamic Therapy for the Treatment of American Tegumentary Leishmaniasis: Evaluation of Therapies Association in Experimentally Infected Mice with Leishmania (Leishmania) amazonensis. J. Lasers Med. Sci. 2018, 9, 274–282. [Google Scholar] [CrossRef]
  136. Cabral, F.V.; Yoshimura, T.M.; Silva, D.F.T.; Cortez, M.; Ribeiro, M.S. Photodynamic Therapy Mediated by a Red LED and Methylene Blue Inactivates Resistant Leishmania amazonensis. J. Opt. Soc. Am. A 2023, 40, 996. [Google Scholar] [CrossRef]
  137. Vannier-Santos, M.; De Castro, S. Electron Microscopy in Antiparasitic Chemotherapy: A (Close) View to a Kill. Curr. Drug Targets 2009, 10, 246–260. [Google Scholar] [CrossRef] [PubMed]
  138. Vannier-Santos, M.A.; Martiny, A.; Lins, U.; Urbina, J.A.; Borges, V.M.; Souza, W. Impairment of Sterol Biosynthesis Leads to Phosphorus and Calcium Accumulation in Leishmania Acidocalcisomes. Microbiology 1999, 145, 3213–3220. [Google Scholar] [CrossRef]
  139. Vannier-Santos, M.A.; Lins, U. Cytochemical Techniques and Energy-Filtering Transmission Electron Microscopy Applied to the Study of Parasitic Protozoa. Biol. Proced. Online 2001, 3, 8–18. [Google Scholar] [CrossRef] [PubMed]
  140. Zhang, K.; Hsu, F.; Scott, D.A.; Docampo, R.; Turk, J.; Beverley, S.M. Leishmania Salvage and Remodelling of Host Sphingolipids in Amastigote Survival and Acidocalcisome Biogenesis. Mol. Microbiol. 2005, 55, 1566–1578. [Google Scholar] [CrossRef] [PubMed]
  141. Klionsky, D.J.; Abdel-Aziz, A.K.; Abdelfatah, S.; Abdellatif, M.; Abdoli, A.; Abel, S.; Abeliovich, H.; Abildgaard, M.H.; Abudu, Y.P.; Acevedo-Arozena, A.; et al. Guidelines for the Use and Interpretation of Assays for Monitoring Autophagy (4th Edition). Autophagy 2021, 17, 1–382. [Google Scholar] [CrossRef] [PubMed]
  142. Onwah, S.S.; Uzonna, J.E.; Ghavami, S. Assessment of Autophagy in Leishmania Parasites. In Methods in Molecular Biology; Springer: New York, NY, USA, 2024; p. 1. [Google Scholar]
  143. Martínez, A.; Carreon, T.; Iniguez, E.; Anzellotti, A.; Sánchez, A.; Tyan, M.; Sattler, A.; Herrera, L.; Maldonado, R.A.; Sánchez-Delgado, R.A. Searching for New Chemotherapies for Tropical Diseases: Ruthenium–Clotrimazole Complexes Display High in vitro Activity against Leishmania major and Trypanosoma cruzi and Low Toxicity toward Normal Mammalian Cells. J. Med. Chem. 2012, 55, 3867–3877. [Google Scholar] [CrossRef]
  144. Fandzloch, M.; Arriaga, J.M.M.; Sánchez-Moreno, M.; Wojtczak, A.; Jezierska, J.; Sitkowski, J.; Wiśniewska, J.; Salas, J.M.; Łakomska, I. Strategies for Overcoming Tropical Disease by Ruthenium Complexes with Purine Analog: Application against Leishmania spp. and Trypanosoma cruzi. J. Inorg. Biochem. 2017, 176, 144–155. [Google Scholar] [CrossRef] [PubMed]
  145. Pereira, J.C.M.; Carregaro, V.; Costa, D.L.; Silva, J.S.; Cunha, F.Q.; Franco, D.W. Antileishmanial Activity of Ruthenium(II)Tetraammine Nitrosyl Complexes. Eur. J. Med. Chem. 2010, 45, 4180–4187. [Google Scholar] [CrossRef]
  146. Holzmuller, P.; Bras-Gonçalves, R.; Lemesre, J.L. Phenotypical Characteristics, Biochemical Pathways, Molecular Targets and Putative Role of Nitric Oxide-Mediated Programmed Cell Death in Leishmania. Parasitology 2006, 132, S19–S32. [Google Scholar] [CrossRef]
  147. Tfouni, E.; Truzzi, D.R.; Tavares, A.; Gomes, A.J.; Figueiredo, L.E.; Franco, D.W. Biological Activity of Ruthenium Nitrosyl Complexes. Nitric Oxide—Biol. Chem. 2012, 26, 38–53. [Google Scholar] [CrossRef]
  148. Orsini, T.M.; Kawakami, N.Y.; Panis, C.; Thomazelli, A.P.F.D.S.; Tomiotto-Pellissier, F.; Cataneo, A.H.D.; Kian, D.; Yamauchi, L.M.; Júnior, F.S.G.; Lopes, L.G.D.F.; et al. Antileishmanial Activity and Inducible Nitric Oxide Synthase Activation by RuNO Complex. Mediat. Inflamm. 2016, 2016, 2631625. [Google Scholar] [CrossRef]
  149. Olekhnovitch, R.; Bousso, P. Induction, Propagation, and Activity of Host Nitric Oxide: Lessons from Leishmania Infection. Trends Parasitol. 2015, 31, 653–664. [Google Scholar] [CrossRef] [PubMed]
  150. Costa, M.S.; Gonçalves, Y.G.; Teixeira, S.C.; Nunes, D.C.O.; Lopes, D.S.; Silva, C.V.; Silva, M.S.; Borges, B.C.; Silva, M.J.B.; Rodrigues, R.S.; et al. Increased ROS Generation Causes Apoptosis-like Death: Mechanistic Insights into the Anti-Leishmania Activity of a Potent Ruthenium(II) Complex. J. Inorg. Biochem. 2019, 195, 1–12. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular structure of methylene blue (MB—(A)), new methylene blue (NMB—(B)), new methylene blue B (NMB-B—(C)), and new methylene blue P (NMB-P—(D)). The structures were constructed using ChemSketch (Freeware) 2023.2.4.
Figure 1. Molecular structure of methylene blue (MB—(A)), new methylene blue (NMB—(B)), new methylene blue B (NMB-B—(C)), and new methylene blue P (NMB-P—(D)). The structures were constructed using ChemSketch (Freeware) 2023.2.4.
Biology 14 01709 g001
Figure 2. In vitro activity of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), new methylene blue P (NMB-P), and miltefosine (MT) on Leishmania amazonensis (106 parasites/mL). Panel A: dose–response curves of MB (A1), NMB (A2), NMB-B (A3), NMB-P (A4), and MT (A5) against promastigote forms (L. amazonensis PH8 strain). Panel B: dose–response curves of MB (B1), NMB (B2), NMB-B (B3), NMB-P (B4), and MT (B5) against ex vivo amastigote forms (L. amazonensis LTB0016 strain). (C,D): Comparison between the compounds and MT at 1.56 μM (C) and 3.12 μM (D) on promastigote forms. (E,F) Comparison between the compounds and MT at <0.10 μM (E) and <0.28 μM (F) on ex vivo amastigote forms. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to negative control (untreated parasites) by one-way ANOVA and Dunnett’s post-test.
Figure 2. In vitro activity of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), new methylene blue P (NMB-P), and miltefosine (MT) on Leishmania amazonensis (106 parasites/mL). Panel A: dose–response curves of MB (A1), NMB (A2), NMB-B (A3), NMB-P (A4), and MT (A5) against promastigote forms (L. amazonensis PH8 strain). Panel B: dose–response curves of MB (B1), NMB (B2), NMB-B (B3), NMB-P (B4), and MT (B5) against ex vivo amastigote forms (L. amazonensis LTB0016 strain). (C,D): Comparison between the compounds and MT at 1.56 μM (C) and 3.12 μM (D) on promastigote forms. (E,F) Comparison between the compounds and MT at <0.10 μM (E) and <0.28 μM (F) on ex vivo amastigote forms. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to negative control (untreated parasites) by one-way ANOVA and Dunnett’s post-test.
Biology 14 01709 g002
Figure 3. In vitro proliferation of Leishmania amazonensis promastigote forms (106 parasites/mL, PH8 strain) exposed to IC50 of methylene blue (MB—61.44 µM), new methylene blue (NMB—5.42 µM), new methylene blue B (NMB-B—5.48 µM), new methylene blue P (NMB-P—2.84 µM), and miltefosine (MT—23.20 µM) over 168 h of incubation. (A) Fluorescence intensity comparison between untreated and treated parasites. (BE) Fluorescence intensity over time for parasites treated with MB (B), NMB (C), NMB-B (D), and NMB-P (E), compared to MT. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to untreated parasites (A) and the standard drug MT (BE) by one-way ANOVA and Dunnett’s post-test. IC50 = 50% Maximal Inhibitory Concentration.
Figure 3. In vitro proliferation of Leishmania amazonensis promastigote forms (106 parasites/mL, PH8 strain) exposed to IC50 of methylene blue (MB—61.44 µM), new methylene blue (NMB—5.42 µM), new methylene blue B (NMB-B—5.48 µM), new methylene blue P (NMB-P—2.84 µM), and miltefosine (MT—23.20 µM) over 168 h of incubation. (A) Fluorescence intensity comparison between untreated and treated parasites. (BE) Fluorescence intensity over time for parasites treated with MB (B), NMB (C), NMB-B (D), and NMB-P (E), compared to MT. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to untreated parasites (A) and the standard drug MT (BE) by one-way ANOVA and Dunnett’s post-test. IC50 = 50% Maximal Inhibitory Concentration.
Biology 14 01709 g003
Figure 4. In vitro cytotoxic activity of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), new methylene blue P (NMB-P), and miltefosine (MT) on mammalian cells after 24 h of incubation. Panel A: L929 cells (0.5 × 105/well) treated with MB (A1), NMB (A2), NMB-B (A3), NMB-P (A4), and MT (A5). Panel B: HepG2 cells (0.5 × 105/well) treated with MB (B1), NMB (B2), NMB-B (B3), NMB-P (B4), and MT (B5). Panel C: VERO cells (105/well) treated with MB (C1), NMB (C2), NMB-B (C3), NMB-P (C4), and MT (C5). Panel D: J774.G8 cells (105/well) treated with MB (D1), NMB (D2), NMB-B (D3), NMB-P (D4), and MT (D5). Panel E: Murine peritoneal macrophages—MPMs (3 × 105/well)—treated with MB (E1), NMB (E2), NMB-B (E3), NMB-P (E4), and MT (E5). Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to positive control (untreated cells) by one-way ANOVA and Dunnett’s post-test.
Figure 4. In vitro cytotoxic activity of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), new methylene blue P (NMB-P), and miltefosine (MT) on mammalian cells after 24 h of incubation. Panel A: L929 cells (0.5 × 105/well) treated with MB (A1), NMB (A2), NMB-B (A3), NMB-P (A4), and MT (A5). Panel B: HepG2 cells (0.5 × 105/well) treated with MB (B1), NMB (B2), NMB-B (B3), NMB-P (B4), and MT (B5). Panel C: VERO cells (105/well) treated with MB (C1), NMB (C2), NMB-B (C3), NMB-P (C4), and MT (C5). Panel D: J774.G8 cells (105/well) treated with MB (D1), NMB (D2), NMB-B (D3), NMB-P (D4), and MT (D5). Panel E: Murine peritoneal macrophages—MPMs (3 × 105/well)—treated with MB (E1), NMB (E2), NMB-B (E3), NMB-P (E4), and MT (E5). Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to positive control (untreated cells) by one-way ANOVA and Dunnett’s post-test.
Biology 14 01709 g004
Figure 5. Mitochondrial membrane potential (ΔΨm) evaluation by flow cytometry on Leishmania amazonensis (5 × 106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h. Panel A (A1A7): evaluation on promastigote forms (L. amazonensis PH8 strain). Panel B (B1B7): evaluation on ex vivo amastigote forms (L. amazonensis LTB0016 strain). (A1,B1) Untreated TMRE+ parasites (negative control). (A2,B2) Untreated TMRE+ and FCCP+ parasites (positive control). (A3,B3) = TMRE fluorescence intensity of negative control. (A4A7,B4B7) TMRE fluorescence intensity of parasites treated with the IC50 of MB (A4,B4), NMB (A5,B5), NMB-B (A6,B6), and NMB-P (A7,B7). (C,D) MFI for promastigotes in LP (C) and HP (D) populations. (E,F) MFI for ex vivo amastigotes in LP (E) and HP (F) populations. Data represent mean ± SD. (*) p < 0.05 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); PE = Channel; TMRE = Tetramethyrhodamine ethyl; FCCP = Carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone; LP = Low Population; HP = High Population; MFI = Median Fluorescence Intensity; IC50 = 50% Maximal Inhibitory Concentration.
Figure 5. Mitochondrial membrane potential (ΔΨm) evaluation by flow cytometry on Leishmania amazonensis (5 × 106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h. Panel A (A1A7): evaluation on promastigote forms (L. amazonensis PH8 strain). Panel B (B1B7): evaluation on ex vivo amastigote forms (L. amazonensis LTB0016 strain). (A1,B1) Untreated TMRE+ parasites (negative control). (A2,B2) Untreated TMRE+ and FCCP+ parasites (positive control). (A3,B3) = TMRE fluorescence intensity of negative control. (A4A7,B4B7) TMRE fluorescence intensity of parasites treated with the IC50 of MB (A4,B4), NMB (A5,B5), NMB-B (A6,B6), and NMB-P (A7,B7). (C,D) MFI for promastigotes in LP (C) and HP (D) populations. (E,F) MFI for ex vivo amastigotes in LP (E) and HP (F) populations. Data represent mean ± SD. (*) p < 0.05 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); PE = Channel; TMRE = Tetramethyrhodamine ethyl; FCCP = Carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone; LP = Low Population; HP = High Population; MFI = Median Fluorescence Intensity; IC50 = 50% Maximal Inhibitory Concentration.
Biology 14 01709 g005
Figure 6. Reactive oxygen species (ROS) production evaluation by flow cytometry on Leishmania amazonensis (5 × 106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h. Panel A (A1A7): evaluation on promastigote forms (L. amazonensis PH8 strain). Panel B (B1B7): evaluation on ex vivo amastigote forms (L. amazonensis LTB0016 strain). (A1,B1) Untreated H2DCFDA+ parasites (negative control). (A2,B2) Untreated H2DCFDA+ and AA+ parasites (positive control for ROS production). (A3,B3) = H2DCFDA fluorescence intensity of negative control. (A4A7,B4B7) H2DCFDA fluorescence intensity of parasites treated with the IC50 of MB (A4,B4), NMB (A5,B5), NMB-B (A6,B6), and NMB-P (A7,B7). MFI in promastigotes HP H2DCFDA+ (C) and ex vivo amastigotes (D) are shown. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); FITC = Channel; H2DCFDA = 2′,7′-dichlorodihydrofluorescein diacetate; AA = Antimycin A; LP = Low Population; HP = High Population; Pro = Promastigotes; Ama = ex vivo Amastigotes; IC50 = 50% Maximal Inhibitory Concentration.
Figure 6. Reactive oxygen species (ROS) production evaluation by flow cytometry on Leishmania amazonensis (5 × 106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h. Panel A (A1A7): evaluation on promastigote forms (L. amazonensis PH8 strain). Panel B (B1B7): evaluation on ex vivo amastigote forms (L. amazonensis LTB0016 strain). (A1,B1) Untreated H2DCFDA+ parasites (negative control). (A2,B2) Untreated H2DCFDA+ and AA+ parasites (positive control for ROS production). (A3,B3) = H2DCFDA fluorescence intensity of negative control. (A4A7,B4B7) H2DCFDA fluorescence intensity of parasites treated with the IC50 of MB (A4,B4), NMB (A5,B5), NMB-B (A6,B6), and NMB-P (A7,B7). MFI in promastigotes HP H2DCFDA+ (C) and ex vivo amastigotes (D) are shown. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); FITC = Channel; H2DCFDA = 2′,7′-dichlorodihydrofluorescein diacetate; AA = Antimycin A; LP = Low Population; HP = High Population; Pro = Promastigotes; Ama = ex vivo Amastigotes; IC50 = 50% Maximal Inhibitory Concentration.
Biology 14 01709 g006
Figure 7. Cell death evaluation by flow cytometry on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with AV and PI (DG). Untreated parasites were used as negative control (AC). The percentage of apoptotic cells (H), necrotic cells (I), and general death (J) are shown. Data represent mean ± SD. (*) p < 0.05 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); FITC and PE = Channels; AV = Annexin V; PI = Propidium Iodide; LQR = Low Right Quadrants; ULRQ = Upper Left and Right Quadrants; IC50 = 50% Maximal Inhibitory Concentration.
Figure 7. Cell death evaluation by flow cytometry on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with AV and PI (DG). Untreated parasites were used as negative control (AC). The percentage of apoptotic cells (H), necrotic cells (I), and general death (J) are shown. Data represent mean ± SD. (*) p < 0.05 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); FITC and PE = Channels; AV = Annexin V; PI = Propidium Iodide; LQR = Low Right Quadrants; ULRQ = Upper Left and Right Quadrants; IC50 = 50% Maximal Inhibitory Concentration.
Biology 14 01709 g007
Figure 8. Cell death evaluation by imaging flow cytometry (Ammnis Stream MK XII) on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with AV and PI. Panel A (A1A4): Negative control (untreated parasites) displayed in brightfield (A1), AV labeling (A2), PI labeling (A3), and merged (A4). Panel B (B1B4): Parasites treated with MB displayed in brightfield (B1), AV labeling (B2), PI labeling (B3), and merged (B4). Panel C (C1C4): Parasites treated with NMB displayed in brightfield (C1), AV labeling (C2), PI labeling (C3), and merged (C4). Panel D (D1D4): Parasites treated with NMB-B displayed in brightfield (D1), AV labeling (D2), PI labeling (D3), and merged (D4). Panel E (E1E4): Parasites treated with NMB-P displayed in brightfield (E1), AV labeling (E2), PI labeling (E3), and merged (E4). AV = Annexin V; PI = Propidium Iodide; IC50 = 50% Maximal Inhibitory Concentration.
Figure 8. Cell death evaluation by imaging flow cytometry (Ammnis Stream MK XII) on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with AV and PI. Panel A (A1A4): Negative control (untreated parasites) displayed in brightfield (A1), AV labeling (A2), PI labeling (A3), and merged (A4). Panel B (B1B4): Parasites treated with MB displayed in brightfield (B1), AV labeling (B2), PI labeling (B3), and merged (B4). Panel C (C1C4): Parasites treated with NMB displayed in brightfield (C1), AV labeling (C2), PI labeling (C3), and merged (C4). Panel D (D1D4): Parasites treated with NMB-B displayed in brightfield (D1), AV labeling (D2), PI labeling (D3), and merged (D4). Panel E (E1E4): Parasites treated with NMB-P displayed in brightfield (E1), AV labeling (E2), PI labeling (E3), and merged (E4). AV = Annexin V; PI = Propidium Iodide; IC50 = 50% Maximal Inhibitory Concentration.
Biology 14 01709 g008
Figure 9. Cell death evaluation by flow cytometry on ex vivo amastigote forms of Leishmania amazonensis LTB0016 strain (10⁶ parasites/mL) exposed to the IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with 7-AAD. (A,B): Negative control (untreated parasites) showing morphology (A) and 7-AAD labeling (B). (CF): Parasites treated parasites with MB (C), NMB (D), NMB-B (E), NMB-P (F), and labeled with 7-AAD. (G): Percentage of parasite death for each treatment compared to negative control. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); PerCP = Channel; 7-AAD = 7-amino-actinomycin D; IC50 = 50% Maximal Inhibitory Concentration.
Figure 9. Cell death evaluation by flow cytometry on ex vivo amastigote forms of Leishmania amazonensis LTB0016 strain (10⁶ parasites/mL) exposed to the IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with 7-AAD. (A,B): Negative control (untreated parasites) showing morphology (A) and 7-AAD labeling (B). (CF): Parasites treated parasites with MB (C), NMB (D), NMB-B (E), NMB-P (F), and labeled with 7-AAD. (G): Percentage of parasite death for each treatment compared to negative control. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001; (***) p < 0.0001 when compared to negative control by one-way ANOVA and Dunnett’s post-test. SSC = Side Scatter (granularity); FSC = Forward Scatter (relative size); PerCP = Channel; 7-AAD = 7-amino-actinomycin D; IC50 = 50% Maximal Inhibitory Concentration.
Biology 14 01709 g009
Table 1. In vitro activities of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), new methylene blue P (NMB-P), and miltefosine on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL), ex vivo amastigote forms of L. amazonensis LTB0016 strain (106 parasites/mL), mammalian cells [L929, HepG2 (0.5 × 105 cells/well), VERO, J774.G8 (105 cells/well), and murine peritoneal macrophages—MPMs (3 × 105 cells/well)]—and their respective selective indexes (SI) after 24 h of incubation.
Table 1. In vitro activities of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), new methylene blue P (NMB-P), and miltefosine on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL), ex vivo amastigote forms of L. amazonensis LTB0016 strain (106 parasites/mL), mammalian cells [L929, HepG2 (0.5 × 105 cells/well), VERO, J774.G8 (105 cells/well), and murine peritoneal macrophages—MPMs (3 × 105 cells/well)]—and their respective selective indexes (SI) after 24 h of incubation.
CompoundsAntiparasitic ActivityCytotoxicity/Selective Indexes
IC50 μMCC50 μM/(SIP/SIA)
Pro.Ama.L929HepG2VEROJ744.G8MPMs
MB61.44 ± 4.4115.65 ± 2.23321.65 ± 3.7495.96 ± 3.28141.05 ± 5.58206.20 ± 9.1936.02 ± 3.69
(5.24/20.55)(1.56/6.13)(2.30/9.01)(3.36/13.18)(0.59/2.30)
NMB5.42 ***
± 0.81
2.66 ± 0.95280.25 ± 2.1971.56 ± 3.1544.73 ± 3.4068.50 ± 2.1231.27 ± 5.72
(51.71/105.35)(13.20/26.90)(8.25/16.82)(12.64/25.75)(5.77/11.76)
NMB-B5.48 ***
± 1.73
3.16 ± 0.37270.30 ± 8.4834.24 ± 7.31247.95 ± 32.88129.80 ± 7.3565.91 ± 1.69
(49.32/85.54)(6.25/10.84)(45.25/78.47)(23.69/41.08)(12.03/20.86)
NMB-P2.84 ***
± 0.80
0.46 ± 0.34150.40 ± 12.0235.05 ± 2.08239.80 ± 27.5769.82 ± 10.3871.63 ± 1.85
(52.96/326.96)(12.34/76.20)(84.44/521.30)(24.58/151.78)(25.22/155.72)
Miltefosine23.20 ± 2.550.97 ± 0.40112.71 ± 18.22181.00 ± 11.31168.05 ± 2.75155.60 ± 8.48102.93 ± 4.77
(4.86/116.20)(7.80/186.60)(7.24/173.25)(6.71/160.41)(4.44/106.11)
Pro. = Promastigotes. Ama. = Amastigotes. IC50 = 50% Maximal Inhibitory Concentration. CC50 = 50% Maximal Cytotoxicity Concentration. Selectivity Index (SI) = CC50/IC50. SIP = Selectivity Index on promastigote forms of L. amazonensis PH8 strain. SIA = Selectivity Index on ex vivo amastigote forms of L. amazonensis LTB0016 strain. Data represent the mean ± SD. (***) p < 0.0001 when compared to the standard drug (MT) by one-way ANOVA and Dunnett’s post-test.
Table 2. Flow cytometry analysis of mitochondrial membrane potential (ΔΨm) on promastigote forms of Leishmania amazonensis PH8 strain (5 × 106 parasites/mL) and ex vivo amastigote forms of L. amazonensis LTB0016 strain (5 × 106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with TMRE.
Table 2. Flow cytometry analysis of mitochondrial membrane potential (ΔΨm) on promastigote forms of Leishmania amazonensis PH8 strain (5 × 106 parasites/mL) and ex vivo amastigote forms of L. amazonensis LTB0016 strain (5 × 106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with TMRE.
CompoundsIC50 (µM)PromastigotesIC50 (µM)ex vivo Amastigotes
Low PopulationHigh PopulationLow PopulationHigh Population
MFIVIΔΨmMFIVIΔΨmMFIVIΔΨmMFIVIΔΨm
MB61.442737.60 *−0.41
± 0.05
56,878.470.02
± 0.03
15.653237.43−0.37
± 0.32
317,106.50−0.61
± 0.18
NMB5.424095.05−0.14
± 0.20
65,590.870.11
± 0.08
2.663770.37−0.27
± 0.06
277,557.80 *−0.43
± 0.20
NMB-B5.484070.50−0.13
± 0.09
74,562.130.27
± 0.16
3.164174.80−0.31
± 0.03
410,112.30−0.59
± 0.21
NMB-P2.843359.03 *−0.28
± 0.03
86,029.230.49
± 0.36
0.467215.470.21
± 0.04
642,051.430.39
± 0.13
Control-4702.030.0059,161.870.00-5226.500.00469,563.430.00
TMRE = Tetramethyrhodamine ethyl. IC50 = 50% Maximal Inhibitory Concentration. MFI = Median Fluorescence Intensity. VIΔΨm (Variation Index of Mitochondrial Membrane Potential) = (MT − MC)/MC, where MT corresponds to the TMRE MFI of treated parasites and MC corresponds to the TMRE MFI of control parasites. The control corresponds to untreated parasites labeled with TMRE. Data represent mean ± SD. (*) p < 0.05 when compared to control by one-way ANOVA and Dunnett’s post-test.
Table 3. Flow cytometry analysis of reactive oxygen species (ROS) production on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL) and ex vivo amastigote forms of L. amazonensis LTB0016 strain (106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with H2DCFDA.
Table 3. Flow cytometry analysis of reactive oxygen species (ROS) production on promastigote forms of Leishmania amazonensis PH8 strain (106 parasites/mL) and ex vivo amastigote forms of L. amazonensis LTB0016 strain (106 parasites/mL) exposed to IC50 of methylene blue (MB), new methylene blue (NMB), new methylene blue B (NMB-B), and new methylene blue P (NMB-P) for 24 h and labeled with H2DCFDA.
CompoundsIC50 (µM)PromastigotesIC50 (µM)Ex Vivo Amastigotes
Low PopulationHigh PopulationLow PopulationHigh Population
MFIVIROSMFIVIROSMFIVIROSMFIVIROS
MB61.4426,534.030.74
± 0.12
1,658,186.80 *3.56
± 0.50
15.653151.501.28
± 0.46
140,636.200.90
± 0.29
NMB5.4231,489.800.87
± 0.12
1,512,521.00 *3.61
± 0.08
2.662940.571.62
± 0.90
131,669.430.84
± 0.51
NMB-B5.4839,191.971.06
± 0.11
2,639,177.00 **5.81
± 0.64
3.162116.871.11
± 0.48
131,896.270.85
± 0.49
NMB-P2.8434,607.801.02
± 0.48
2,325,902.50 **5.39
± 1.83
0.463063.171.27
± 0.45
140,291.570.90
± 0.45
AA1019,775.400.52
± 0.13
890,705.802.07
± 0.23
103011.231.14
± 0.38
143,227.250.60
± 0.53
Control-39,868.50-452,909.35--2436.40-158,240.00-
H2DCFDA = 2′,7′-dichlorodihydrofluorescein diacetate. IC50 = 50% Maximal Inhibitory Concentration. AA = Antimycin A. MFI = Median Fluorescence Intensity. VIROS (Variation Index of ROS production) = MT/MC, where MT corresponds to the H2DCFDA MFI of treated parasites, and MC corresponds to the H2DCFDA MFI of control parasites. The control corresponds to untreated parasites labeled with H2DCFDA. Data represent mean ± SD. (*) p < 0.05; (**) p < 0.001 when compared to negative control by one-way ANOVA and Dunnett’s post-test.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vasco-dos-Santos, D.R.; Almeida-Silva, J.; Fiuza, L.F.d.A.; Vacani-Martins, N.; Rocha, Z.N.d.; Soeiro, M.d.N.C.; Henriques-Pons, A.; Torres-Santos, E.C.; Vannier-Santos, M.A. From Dyes to Drugs? Selective Leishmanicidal Efficacy of Repositioned Methylene Blue and Its Derivatives in In Vitro Evaluation. Biology 2025, 14, 1709. https://doi.org/10.3390/biology14121709

AMA Style

Vasco-dos-Santos DR, Almeida-Silva J, Fiuza LFdA, Vacani-Martins N, Rocha ZNd, Soeiro MdNC, Henriques-Pons A, Torres-Santos EC, Vannier-Santos MA. From Dyes to Drugs? Selective Leishmanicidal Efficacy of Repositioned Methylene Blue and Its Derivatives in In Vitro Evaluation. Biology. 2025; 14(12):1709. https://doi.org/10.3390/biology14121709

Chicago/Turabian Style

Vasco-dos-Santos, Deyvison Rhuan, Juliana Almeida-Silva, Ludmila Ferreira de Almeida Fiuza, Natalia Vacani-Martins, Zênis Novais da Rocha, Maria de Nazaré Correia Soeiro, Andrea Henriques-Pons, Eduardo Caio Torres-Santos, and Marcos André Vannier-Santos. 2025. "From Dyes to Drugs? Selective Leishmanicidal Efficacy of Repositioned Methylene Blue and Its Derivatives in In Vitro Evaluation" Biology 14, no. 12: 1709. https://doi.org/10.3390/biology14121709

APA Style

Vasco-dos-Santos, D. R., Almeida-Silva, J., Fiuza, L. F. d. A., Vacani-Martins, N., Rocha, Z. N. d., Soeiro, M. d. N. C., Henriques-Pons, A., Torres-Santos, E. C., & Vannier-Santos, M. A. (2025). From Dyes to Drugs? Selective Leishmanicidal Efficacy of Repositioned Methylene Blue and Its Derivatives in In Vitro Evaluation. Biology, 14(12), 1709. https://doi.org/10.3390/biology14121709

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

Article Metrics

Back to TopTop