Hostname: page-component-669899f699-chc8l Total loading time: 0 Render date: 2025-04-25T23:49:53.520Z Has data issue: false hasContentIssue false

Plant-derived products as anti-leishmanials which target mitochondria: a review

Published online by Cambridge University Press:  26 March 2025

Chandrima Shaha*
Affiliation:
Division of Infectious Diseases and Immunology, Indian Institute of Chemical Biology, Kolkata, India
*
Corresponding author: Chandrima Shaha; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Background

The global incidences of leishmaniasis are increasing due to changing environmental conditions and growing poverty. Leishmaniasis, caused by the Leishmania parasite, presents itself in six different clinical forms, the cutaneous and the visceral diseases being the most prevalent. While the cutaneous form causes disfigurement, the visceral form could be fatal if not treated. With no available vaccines combined with serious side effects of current medications and emerging drug resistance, it is crucial to discover new drugs whether as novel compounds or as repurposed existing pharmaceuticals. In the realm of drug development, mitochondria are recognized as important pharmacological targets due to their critical role in energy control, which, when disrupted, leads to irreversible cell damage. Certain plant-based compounds able to target the parasite mitochondrion, have been studied for their potential anti-leishmanial effects.

Search results

These compounds have shown promising effects in eliminating the Leishmania parasite. Artemisinin and chloroquine, two anti-malarial drugs that target mitochondria, exert strong anti-leishmanial effectiveness in both in vitro cultures and in vivo animal models. Quinolones, coumarins and quercetin are other compounds with leishmanicidal properties, which disrupt mitochondrial activity to effectively eliminate parasites in animal models of the disease and could be considered as potential drugs.

Conclusions

Therefore, plant-based compounds hold promise as potential candidates for anti-leishmanial drug development.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2025. Published by Cambridge University Press

Introduction

A considerable section of the global population uses plant-based medicines to cure ailments, and about 75% of medications created to treat infectious diseases have their roots in nature (Refs Reference Mathur and Hoskins1Reference Theodoridis, Drakou, Hickler, Thines and Nogues-Bravo3). In light of the above findings, the search for medications derived from plants to treat neglected tropical illnesses continues. Recent technological advancements give access to a vast array of novel skeletons from plant sources where combinatorial modification may be able to yield suitable non-toxic products (Ref Reference Alcazar, Alakurtti, Padrón-Nieves, Tuononen, Rodríguez, Yli-Kauhaluoma and Ponte-Sucre4, Reference Atanasov, Zotchev, Dirsch, Supuran and Taskforce5). Leishmaniasis is one of the seven most serious tropical diseases with 700,000 to 1 million new cases reported annually (Ref 6). The current chemotherapy for leishmaniasis is not efficient due to long administration schedules, resistance development, high cost, clinical failures and toxicity, so the development of new treatments has been prioritized (Ref Reference Ghorbani and Farhoudi7). To achieve the elimination of leishmaniasis, the discovery of new anti-leishmanial compounds or repurposing of existing drugs is necessary (Refs Reference Charlton, Rossi-Bergmann, Denny and Steel8, Reference Bustamante, Ochoa, Asela and Muskus9). This focused review attempts to identify available plant-based compounds that target the parasite’s mitochondrion to disrupt cellular function and have been tested in the human or animal system.

Search strategy

Electronic databases of PubMed and Google Scholar were used with ‘plants’ and ‘Leishmania’ as keywords for a period span of years 1985–2024. About 1718 results were listed. From this, the selection of articles was performed based on the title and subsequently the evaluation of the abstract for eligibility, followed by an assessment of the full text for relevance. From here, 160 references were extracted and further analyzed, according to the inclusion/study selection criteria for plant extracts having good anti-leishmanial effects affecting the mitochondria and already tested in humans or animals. This forms the primary body of the review. A separate section at the end evaluates some possible compounds that have been only tested in vitro but target the mitochondria of the parasite. This information may form a basis for future explorations in suitable models.

Leishmaniasis and the Leishmania parasite

Leishmaniasis, caused by the parasite Leishmania manifests in six clinical forms, namely, visceral leishmaniasis (VL), post-kala-azar dermal leishmaniasis (PKDL), cutaneous leishmaniasis (CL), diffuse cutaneous leishmaniasis (DCL), mucocutaneous leishmaniasis (MCL) and mucosal leishmaniasis (ML)(Ref Reference Akhoundi, Downing, Votýpka, Kuhls, Lukeš, Cannet and Sereno10). Over the past few years, zoonotic or anthroponotic transmissions have caused the re-emergence of VL and CL because of political instability, deforestation and climate change (Refs Reference Hotez11Reference Al-Salem, Pigott, Subramaniam, Haines, Kelly-Hope, Molyneux and Acosta-Serrano13). VL is the second most lethal parasitic disease after malaria and is fatal in the absence of therapy (Ref Reference Torres-Guerrero, Quintanilla-Cedillo, Ruiz-Esmenjaud and Arenas14). VL can also present as PKDL, a cutaneous variant of the illness, which presents a risk of recurrence because people with PKDL act as reservoirs for the parasite that causes VL (Ref Reference Kumar, Singh, Tiwari, Madhukar, Rajneesh and Kumar15). Different species of the parasite are distributed all over the world and for reference are designated as the Old World species and the New World species (Ref Reference Kevric, Cappel and Keeling16) (Figure 1).

Figure 1. Distribution of leishmaniasis in the Old World and the New World. The figure shows species distribution of the Leishmania parasite in the old world and the new world (Ref Reference Kevric, Cappel and Keeling16). CL: cutaneous leishmaniasis; MCL: mucocutaneous leishmaniasis; VL: visceral leishmaniasis.

Chemotherapy with pentavalent antimonials as sodium stibogluconate, has been used as a first-line drug against VL since 1945 and is still being used for the treatment of canine leishmaniasis (Ref Reference Miret, Moreno, Nieto, Carter, Mullen, Ambros and González17). The use of antimony in humans is limited because of the development of resistance, high levels of toxicity and occasionally therapeutic failure (Ref Reference Olías-Molero, de la Fuente, Cuquerella, Torrado and Alunda18). Amphotericin B (AmB), a polyene antibiotic is used as an anti-leishmanial which is more effective in lipidic presentations (Ref Reference Olías-Molero, de la Fuente, Cuquerella, Torrado and Alunda18) but high cost and toxicity prevent its widespread use. Oral drug Miltefosine is a neoplastic agent that has been used as an anti-leishmanial but teratogenic effects have limited its use (Ref Reference Sundar and Olliaro19). An aminoglycoside antibiotic, paromomycin, was also employed as an anti-leishmanial medication, but its usage was restricted due to the quick emergence of resistance (Ref Reference Jhingran, Chawla, Saxena, Barrett and Madhubala20). Pentamidine showed high toxicity when used against VL and is now used mostly for CL (Ref Reference Olías-Molero, de la Fuente, Cuquerella, Torrado and Alunda18).

The Leishmania parasite exhibits two morphological forms during its digenetic life cycle in two different hosts, a mammalian host and an invertebrate host the sand fly (Figure 2). Humans, dogs and rats are the mammalian hosts, while the invertebrate host, the sand-fly belongs to either the Phlebotomus or Lutzomyia genus. Parasites picked up from infected mammals grow in the sand fly gut as free-swimming forms (promastigotes) at a temperature of about 25°C and upon maturity move to the proboscis of the fly as infective metacyclic promastigotes (Ref Reference Torres-Guerrero, Quintanilla-Cedillo, Ruiz-Esmenjaud and Arenas14). These free-swimming metacyclics are introduced into the skin of the mammalian host (37°C) through the sand fly bite, from where they are picked up by the circulating phagocytes for elimination. Eventually, the parasites that survive the host defense processes, establish themselves within the phagolysosomes of macrophages, after undergoing major changes in morphology as stationary amastigotes (Ref Reference Clos, Grünebast and Holm21). These amastigotes are the disease-causing forms.

Figure 2. Life cycle of the Leishmania parasite. (1) Sandfly, the invertebrate host, releases infective metacyclic promastigotes in the mammalian bloodstream while taking a blood meal. The parasites enter an environment of 37°C in the mammalian skin from where they are picked up by the phagocytic cells. (2) The promastigotes convert to amastigotes and proliferate within the phagolysosomes of the mammalian macrophages. (3) Amastigotes are released from the macrophages when saturating numbers are reached within a cell. (4) Amastigotes and parasitized macrophages are picked up by the sandfly during a bite. (5) Amastigotes convert to promastigotes within the gut of the sandfly and the infective metacyclic forms move to the proboscis to be delivered to the mammalian bloodstream during a bite (Ref Reference Torres-Guerrero, Quintanilla-Cedillo, Ruiz-Esmenjaud and Arenas14).

Models for testing of anti-leishmanial agents

In vitro cultures for testing anti-leishmanial compounds

Anti-leishmanial drugs are tested in vitro, using both the intracellular amastigotes and the free-swimming promastigotes. For culturing Leishmania parasites from cutaneous leishmaniasis patients, parasites are recovered from a needle biopsy sample of the skin of an infected person, and after purification, parasites are put in culture either in blood agar slants or in enriched media (Ref Reference Paun22). For collecting specimens for culture for the parasites causing the visceral form of the disease, bone marrow, spleen, liver, lymph node or other tissue biopsies are taken and put in culture either in blood agar slants or in enriched media. After incubation, parasites are retrieved (Ref Reference Sundar and Rai23).

The promastigotes divide rapidly in vitro in the logarithmic phase which transforms into stationary phases from late log phase promastigotes and are highly infective (Ref Reference Arjmand, Madrakian, Khalili, Najafi Dastnaee, Zamani and Akbari24). Amastigotes obtained from human biopsies or infected animals or macrophages infected in vitro (Ref Reference Sudhandiran and Shaha25) are used for experiments. Axenic amastigotes, a type of amastigote commonly employed in in vitro research, are generated from promastigotes cultivated at 37°C and pH 5.0, which replicates the conditions of a phagolysosome of the host macrophage that the parasite uses as the survival niche (Ref Reference Chanmol, Jariyapan, Somboon, Bates and Bates26).

Typically, three methods are employed for in vitro experiments with amastigotes. First, mammalian macrophage cultures are infected with promastigotes of Leishmania spp. These parasites will transform into amastigotes between 3 and 5 days (depending on the species of the parasite). The parasites are then collected from the macrophages by differential centrifugation (30 g for macrophages and 700 g for the parasites). The second technique is the use of axenic amastigotes in vitro, generated by the method stated in the earlier paragraph. The third possible way is to collect amastigotes from skin lesions or liver biopsies and use them for in vitro testing.

In vivo testing for anti-leishmanial compounds

For in vivo animal testing, mice or hamsters are the most common models while dog and non-human primates are used less frequently (Ref Reference Loría-Cervera and Andrade-Narváez27). Several Leishmania species like Leishmania major (L. major), Leishmania mexicana (L. Mexicana) and Leishmania amazonensis (L. amazonensis) are used to generate CL (Ref Reference Loría-Cervera and Andrade-Narváez27) by injection in the foot pad of the animal. Sensitive BALB/c mice get extensive lesions that lead to the progression of the illness and serve as models for the study of CL (Ref Reference Loría-Cervera and Andrade-Narváez27).

For generating VL, mice are challenged intravenously with late log or stationary phase promastigotes of Leishmania infantum (L. infantum) or Leishmania donovani (L. donovani). Two to 4 weeks after injection, amastigotes can be recovered from the spleen and liver (Ref Reference Shivahare, Vishwakarma, Parmar, Yadav, Haq, Srivastava and Kar28). The most suitable model for visceralizing Leishmania infections is the Syrian golden hamster (Mesocricetus auratus) (Ref Reference Shivahare, Vishwakarma, Parmar, Yadav, Haq, Srivastava and Kar28). A canine model and the Asian rhesus macaques (Macaca mulatta) are quite susceptible to the Leishmania infection and are used as disease models (Ref Reference Loría-Cervera and Andrade-Narváez27).

The Leishmania mitochondrion and the electron transport chain

There are prominent differences between the Leishmania mitochondrion and the mammalian mitochondria. In terms of numbers, there is a single long-branching mitochondrion in the Leishmania cells that extends throughout the body (Ref Reference Kathuria, Bhattacharjee, Sashidhara, Singh and Mitra29). In contrast, there are smaller mitochondria in large numbers that populate the mammalian cells. In the parasite, this organelle contains a dense complex of DNA near the basal body towards the flagellar end called the kinetoplast which is not present in mammalian mitochondria. Unlike the mammalian mitochondrial DNA, the Leishmania mitochondrial DNA (mtDNA) is made up of thousands of minicircles and some maxicircles that form a complex network (Ref Reference Camacho, Rastrojo, Sanchiz, González-de la Fuente, Aguado and Requena30). The mitochondrial electron transport chain is an important target as interference with its function impacts the survival of the parasite. The cytochrome bc1 complex is one such target. In addition, the protein-importing machinery of the Leishmania mitochondria is a possible target, like the specific protein pATOM36 (peripheral atom 36), the archaic translocase of the outer mitochondrial membrane is not present in mammalian cells (Ref Reference Pusnik, Mani and Schmidt31). In addition, defense proteins like the mitochondrial tryparedoxin peroxidase in the mitochondria, unique to the parasite would be specific targets.

Existing drugs like amphotericin B, miltefosine, pentamidine and antimonials interfere with the mitochondrial membrane permeability of the parasite, resulting in a collapse of the mitochondrial membrane potential (ΔΨm) (Refs Reference Mehta and Shaha32Reference Shadab, Jha, Asad, Deepthi, Kamran and Ali34). Given the susceptibility of the parasite mitochondria to external agents, it is a good drug target. In general, mitochondria are recognized as targets for drugs (Ref Reference Zinovkin and Zamyatnin35) because they control the most important aspects of cell viability like the energy balance, essential for homeostasis and survival. Since any interference with energy homeostasis would be an ideal target, in the last decade, many mitochondria-targeted drugs have been synthesized and tested in the mammalian system and some are in clinical trials (Ref Reference Bastidas, Araya-Valdés, Cortés, Jara and Catalán36Reference Monzote and Gille38). These drugs, already found effective in the mammalian system, should be tested as anti-leishmanial agents in both in vitro and in vivo studies. In this review, compounds affecting the parasite mitochondria have been highlighted with the expectation that this information will serve as a base for designing new molecules or repurposing existing drugs.

The mitochondrial respiratory chain of parasites shows greater diversity in the electron transport complexes (ETC) than their hosts making them good targets for therapy (Ref Reference Zorova, Popkov, Plotnikov, Silachev, Pevzner, Jankauskas and Zorov39). The ETC consists of integral membrane proteins like Complex I (NADH ubiquinone oxidoreductase), Complex II (succinate ubiquinone oxidoreductase), Complex III (cytochrome c3+ oxidoreductase) and Complex IV (cytochrome C oxidase with ubiquinone) localized on the inner mitochondrial membrane (Figure 3). The equilibrium of NADH+/NAD is preserved by the ETC, which is essential for cell viability. Complexes I, III and IV function as proton pumps by allowing protons to move from the matrix into the intermembrane space in opposition to the gradient (Figure 3). The pumping of protons results in the generation of an electrochemical gradient between the intermembrane space and the mitochondrial matrix. The ΔΨm thus produced, drives ATP synthesis from ADP and inorganic phosphate by the F1F0 ATP synthase (Ref Reference Zorova, Popkov, Plotnikov, Silachev, Pevzner, Jankauskas and Zorov39). The ΔΨm is the main component of the electrochemical H+ gradient and is a dynamic measure of mitochondrial function. A break in the electron transport chain’s operation, ATP depletion, loss of ΔΨm, ROS generation or an increase in intracellular or mitochondrial Ca2+ could all lead to the functional disruption of mitochondria (Ref Reference Ly, Grubb and Lawen40). The ΔΨm plays a central role in mediating the apoptotic process (Ref Reference Menna-Barreto and de Castro41), but the alteration of ΔΨm can also occur secondary to (i) inhibition of the ETC, (ii) stimulation of uncoupling proteins and/or (iii) permeabilization of the inner membrane. Blocking the ETC can generate toxic reactive oxygen species (ROS) such as superoxide anion (O2), and uncoupling of the ETC in kinetoplastid parasites was shown to increase the generation of toxic ROS, with complex III, also known as cytochrome bc1, being the principal ROS source (Ref Reference Menna-Barreto and de Castro41). Therefore, the delivery of drugs to the mitochondrion of Leishmania could be designed to inhibit the mitochondrial permeability transition (MPT), uncouple the electron transport chain (ETC) or activate the uncoupling proteins.

Figure 3. The electron transport chain. Complex I (NADH ubiquinone oxidoreductase), complex II (succinate ubiquinone oxidoreductase); complex III (cytochrome c3+ oxidoreductase); complex IV (cytochrome C oxidase with ubiquinone) are located on the inner mitochondrial membrane as integral membrane proteins. Complex I transfers two electrons to ubiquinone when a simultaneous translocation of protons occurs. Coenzyme Q and cytochrome C serve as mobile electron carriers to facilitate the production of ATP through oxidative phosphorylation. CoQ10 allows the transfer of electrons to complex III which transfers these electrons to the cytochrome C responsible for connecting to complex IV where the reduction of O2 to H2O will take place. Complexes I, III, and IV function as proton pumps. The generation of an electrochemical gradient between the intermembrane space and the mitochondrial matrix occurs because of the pumping of protons. The ΔΨm thus produced drives ATP synthesis from ADP and inorganic phosphate by the F1F0 ATP synthase (Refs Reference Zinovkin and Zamyatnin35, Reference Bastidas, Araya-Valdés, Cortés, Jara and Catalán36). Cyt C: cytochrome c; IMM: inner mitochondrial membrane; IMS: Intermembrane space; OMM: Outer mitochondrial membrane; NADH: nicotinamide adenine dinucleotide + hydrogen.

The Leishmania parasite while infecting the host, faces severe oxidative stress from the cells of the host immune system. Particularly, it is a disadvantage that the parasite lacks catalase which is essential to eliminate reactive oxygen species, and therefore, Leishmania has adapted to use a different enzyme system, the tryperodoxin peroxidases. These enzymes are located in the cytosol and the mitochondria and are used by the parasite to eliminate hydrogen peroxide and products generated by it (Ref Reference Das, Giri, Sundar and Shaha42).

Plant-derived natural products as anti-leishmanial agents targeting the mitochondrion

Artemisinin

Artemisinin, a sesquiterpene lactone, and its derivatives have been widely used for malaria. Based on references to the use of the herb Artemisia annua in Chinese traditional medicine, artemisinin was discovered by Tu Youyou in 1972 (Ref Reference Wang, Xu, Wong, Liao, Jiang and Tu43). Through a collaborative effort, Chinese scientists prepared dihydroartemisinin, artemether and artesunate generically known as ‘artemisinins’ from Artemisia annua that were used as anti-malarial medications (Ref Reference Wang, Xu, Wong, Liao, Jiang and Tu43). WHO recommends artemisinin-based combination therapies as the second line of treatment for P. vivax that are resistant to chloroquine and the first line of treatment for uncomplicated P. falciparum malaria (Ref Reference Kong and Tan44). As a remedy for malaria in many endemic areas, artemisinin derivatives such as artesunate and artether are used solo or in combination (Ref Reference Chen and Hsiang45, Reference Krishna, Bustamante, Haynes and Staines46). Artemisinin is a tetracyclic 1,2,4-trioxane containing an endoperoxide bridge (C–O–O–C), the key pharmacophore of the drug. (Figure 4). Notably, it is not only malaria that artemisinin is used for, but it is also prescribed for phylogenetically unrelated parasitic infections of public health importance like schistosomiasis (Ref Reference Issa, Warsame, Mahamat, Saleh, Boulotigam, Djimrassengar and Menard47) or other helminthic diseases caused by nematodes, trematodes, cestodes and others (Ref Reference Salas-Coronas, Vázquez-Villegas, Villarejo-Ordóñez, Sánchez-Sánchez, Espada-Chavarría, Soriano-Pérez and Cabezas-Fernández48Reference Bergquist and Elmorshedy50). Therefore, this drug has a wide range of efficacy against parasites which makes it suitable for repurposing.

Figure 4. In vivo tested plant-based possible anti-leishmanials that could target mitochondria. Artemisinin, chloroquine, quercetin, coumarin and R1–R5 quinolone are shown.

In an in vitro and an ex-vivo study for VL, apoptotic death of L. donovani promastigotes and intracellular amastigotes was induced by artemisinin with IC50 values of 160 and 22 μM, respectively (Ref Reference Sen, Bandyopadhyay, Dutta, Mandal, Ganguly, Saha and Chatterjee51). A loss of mitochondrial potential occurred before initiation of the death process (Ref Reference Sen, Bandyopadhyay, Dutta, Mandal, Ganguly, Saha and Chatterjee51). Other studies confirmed similar observations in in vitro and ex-vivo studies showing leishmanicidal activity against L. donovani with 50 % inhibitory concentration of 14.63 ± 1.49 μg ml−1 and 7.3 ± 1.85 μg ml−1, respectively, against the promastigotes and intracellular amastigotes. Reactive oxygen species (ROS) and nitric oxide NO were shown to precipitate mitochondrial changes leading to parasite death by apoptosis (Ref Reference Islamuddin, Chouhan, Tyagi, Abdin, Sahal and Afrin52). The leishmanicidal activity shown in in vitro and ex-vivo experiments was supported by in vivo studies in a model of leishmaniasis in infected BALB/c mice where the extracts were 90% effective in eliminating parasites (Ref Reference Islamuddin, Chouhan, Tyagi, Abdin, Sahal and Afrin52). Artemisinin incorporated in liposomes with increased availability demonstrated a noteworthy anti-leishmanial efficacy both in vivo in L. donovani-infected BALB/c mice and ex vivo in murine macrophages (Ref Reference Want, Islammudin, Chouhan, Ozbak, Hemeg, Chattopadhyay and Afrin53, Reference Aderibigbe54). In the BALB/c mice, the efficacy was excellent with percentage inhibition of the parasite load of 82.4% ± 3.8% in the liver and 77.6% ± 5.5% in the spleen (Ref Reference Aderibigbe54). Already tested in humans for safety (Ref Reference Li, Guo, Fu, Jian and Wang55), artemisinin and its derivatives or the combinations could be considered as potential anti-leishmanials.

For CL, intracellular amastigotes of L. amazonensis, prevalent in the Brazilian Amazon region and capable of causing CL and MCL, were eliminated when exposed to artemisinin, artemether, artesunate and dihydroartemisinin (Ref Reference Machín, Nápoles, Gille and Monzote56). The in vitro studies were supported by in vivo observations of CL in BALB/c mice created by L. amazonensis where artemisinin and artesunate could reduce parasite load and lesion size (Ref 56). Artesunate, a more stable artemisinin derivative effective in eliminating L. amazonensis parasites, also reduced lesion size and associated inflammation (Ref Reference Gugliandolo, Palma, Peritore, Siracusa, D’Amico, Fusco and Crupi57). L. major, another species of Leishmania parasite causing CL was also eliminated from skin lesions (Ref Reference Ghaffarifar, Esavand Heydari, Dalimi, Hassan, Delavari and Mikaeiloo58). The efficacy shown in vivo generates possibilities for artemisinin and its derivatives as possible drugs for CL as well.

Mechanistically, artemisinin inhibits complexes I–III and II–III of the Leishmania ETC in vitro rapidly increasing ROS, thus disrupting mitochondrial function (Ref Reference De Sarkar, Sarkar, Sarkar, Dighal, Chakrabarti, Staniek and Chatterjee59). The endoperoxide 1,2,4-trioxane ring in artemisinin is responsible for the action of artemisinin against malaria (Ref Reference Wang, Huang, Li, Fan, Long, Li and Zhou60). Structurally, the unusual peroxide bridge of artemisinin in the presence of reducing agents such as Fe2+, haem and Cu2+ becomes unstable by cleavage, a cause of the generation of carbon and oxygen-centred free radicals (Ref Reference Geroldinger, Tonner, Hettegger, Bacher, Monzote, Walter and Gille61). The suppression of ETC is a major source of ROS (Ref Reference Staniek, Gille, Kozlov and Nohl62) especially superoxide. The resulting mitochondrial malfunction affects the energy needs of Leishmania parasites, with ATP depletion causing an energy catastrophe that ultimately results in death (Ref Reference De Sarkar, Sarkar, Sarkar, Dighal, Chakrabarti, Staniek and Chatterjee59). Data on mitochondrial ΔΨm in the Leishmania parasite post artemisinin treatment corroborates observations of the collapse of the ΔΨm in malaria parasites (Ref Reference Antoine, Fisher, Amewu, O’Neill, Ward and Biagini63). The respiratory control rate which is a measure of functioning of the mitochondrial ETC, is reduced by 30% when dihydroartemisinin is added to L. braziliensis cultures, suggesting a mitochondria-targeted action (Ref Reference Grazzia, Boaventura, Garcia, Gadelha and Miguel64). The above studies give a clear indication that the primary site of artemisinin action in the parasite is the mitochondria and with its anti-leishmanial efficacy could be considered for therapy against leishmaniasis.

Chloroquine

Encouraged by the scaffold of quinine, chloroquine, an alkaloid, was first synthesized by chemists in 1934 in Germany (Ref Reference Zhou and Yue65, Reference Frosch, Schmitt, Bringmann, Kiefer and Popp66). It is an analog of quinoline with an aliphatic amino side chain at the 4-position and a chloro group at the 7-position (Ref Reference Frosch, Schmitt, Bringmann, Kiefer and Popp66)(Figure 4). Chloroquine was the drug of choice for malaria till new antimalarials like artemisinin, mefloquine and pyrimethamine were developed. Since then, chloroquine and its derivatives have been repurposed for HIV, systemic lupus erythematosus, rheumatoid arthritis and several other diseases (Ref Reference Carmo, Silva, Machado, Fontes, Pavan, Leite and Da Silva67, Reference Pandey, Sharma, Pandey, Alam, Shaquiquzzaman and Akhter68) and are being tested for anti-tumor therapies based on their action on the autophagic activities of the cell (Ref Reference Ferreira, Soares, Barreto-Junior, Nascimento, Freire-de-Lima, Delorenzi and Pinto-da-Silva69). The distribution of its range of action prompted scientists to test chloroquine and its derivatives for anti-leishmanial efficacy.

Quinolinyl β-enaminone derivatives showed leishmanicidal activity against L. donovani by impairing the mitochondrial ETC complex and inducing ROS-mediated programmed cell death with 84% clearance of parasites in infected mice and hamsters (Ref Reference Rani, Khanikar, Dutta, Katiyar, Qamar, Seth and Kar70). The addition of small heterocyclic rings on the pendent nitrogen of the 4-amino-7-cholroquinoline with replacement of the 7-chloro group with a trifluoromethyl group showed 81-99% inhibition at 10 μM concentration of promastigotes in in vitro cultures. The majority of the analogs demonstrated in vitro anti-amastigote action (IC50 = 5.3–9.6 μM). The substances that demonstrated anti-amastigote activity in vitro were then examined in vivo on a hamster model of VL generated by L. donovani. At 27 days after infection, the 2,6-trichlorophenylthiazolidin-4-one molecule outperformed others, showing around 80% elimination of the Leishmania parasite in treated hamsters (Ref Reference Singh, Kashif, Srivastava and Manna71). One benefit of these compounds is that they are well-tolerated by humans and the thiazolidin/thiazinan-4-ones are biologically advantageous scaffolds (Ref Reference Verma and Saraf72). Other studies reported the identification of two 4-amino-7-chloroquinoline compounds with adamantane or benzothiophene moieties being highly effective as anti-leishmanial candidates in a mouse model of VL using L. infantum. Parasite clearance from the liver was seen at 95% and 99% for doses of 50 and 100 mg/kg, respectively (Ref Reference Konstantinović, Videnović, Orsini, Bogojević, D’Alessandro, Scaccabarozzi and Šolaja73). Interestingly, Sitamaquin, an 8-aminoquinoline analogue, demonstrated superior anti-leishmanial effectiveness in vivo (Ref Reference Loiseau, Cojean and Schrével74). This orally administered 8-amino-quinoline showed efficacy in various animal models of leishmaniasis and encouraging efficacy against various species of Leishmania was found in preliminary clinical studies in Kenya and Brazil (Ref Reference Jha, Sundar, Thakur, Felton, Sabin and Horton75). Nitroimidazo-oxazole compound DNDI-VL-2098 was previously identified as a favourable candidate but was found to cause organ toxicity in animal studies of longer duration and hence was not taken further (Ref Reference Gupta, Yardley, Vishwakarma, Shivahare, Sharma, Launay and Puri76). However, its oxazine derivative DNDI-0690 was recognized as a promising candidate for a Phase I trial for VL (Ref Reference Van Bocxlaer, McArthur, Harris, Alavijeh, Braillard, Mowbray and Croft77) and also for CL (Ref Reference Wijnant, Croft, de la Flor, Alavijeh, Yardley, Braillard and Van Bocxlaer78). The added advantage of the above compound is its efficacy against CL in addition to VL. In 2022, the evaluation of the safety and pharmacokinetic properties of DNDI-0690 was completed. The drug was advanced for consideration for efficacy testing in leishmaniasis patients (Ref Reference Antinarelli, Dias, Souza, Lima, Gameiro, da Silva and Coimbra79). Therefore, the above compounds or their modified versions offer possibilities for treatment of VL.

The 4,7-dichloroquinoline linked together with diamines, diaminealkynes and diaminedialkynes, a series of 4-amino-7-chloroquinoline derivatives showed strong anti-leishmanial activity against L. chagasi, L. braziliensis, L. major and L. amazonensis all causing CL (Ref Reference Carmo, Silva, Machado, Fontes, Pavan, Leite and Da Silva67). These studies again suggested that modification at the 7-position is an attractive strategy for the development of anti-leishmanial compounds. Synthesized 4-aminoquinoline derivatives through conjugation of sulfonamide, hydrazide and hydrazine residues were effective in killing L. amazonensis promastigotes and intracellular amastigotes (Ref Reference Herrera, Llanes, Álvarez, Degracia, Restrepo, Rivera and Fernández80) showing their activity against CL. A chloroquine analog tested on L. panamensis causing tegumentary leishmaniasis showed good efficacy in an in vivo model of BALB/c mice but no mitochondrial interference was checked (Ref Reference Carvalho, Luque-Ortega, Manzano, Castanys, Rivas and Gamarro81).

Mechanistically, aminoquinolines particularly target mitochondrial complex enzymes to disrupt the electron transport system in the mitochondria of protozoan parasites like Plasmodium, Leishmania, etc. (Ref Reference Carvalho, Luque-Ortega, López-Martín, Castanys, Rivas and Gamarro82). Sitamaquin inhibits complex II of the respiratory chain of L. donovani promastigotes (Ref Reference Wang, Sheaff and Hussaini83) where oxidative stress is generated because the drug targets succinate dehydrogenase of complex II. Tafenoquine, an antiplasmodial 8-aminoquinoline, targets Leishmania respiratory Complex III and induces apoptosis (Ref Reference Carvalho, Luque-Ortega, López-Martín, Castanys, Rivas and Gamarro82). Exposure to 7-chloro-4-quinolinylhydrazone derivative to Leishmania induces a loss of mitochondrial potential and generates ROS leading to cell death (Ref Reference Herrera, Llanes, Álvarez, Degracia, Restrepo, Rivera and Fernández80). Chloroquine derivatives are also important disruptors of mitochondrial function through ATP inhibition because any interference with ETC would disrupt ATP (Ref Reference Wang, Sheaff and Hussaini83).

Coumarins

Coumarins are a naturally occurring class of phenolic compounds of the benzopyrone family first isolated from the plant named Dipteryx odorata Willd. by Vogel in 1820 (Ref Reference Küpeli Akkol, Genç, Karpuz, Sobarzo-Sánchez and Capasso84). Coumarins have a wide variety of structures with coumarin 7 as the privileged scaffold (Ref Reference Stefanachi, Leonetti, Pisani, Catto and Carotti85). They are aromatic organic chemical compounds that could be described as having a benzene molecule with two adjacent hydrogen atoms replaced by an unsaturated lactone ring forming a 6-membered heterocycle that shares two carbons with the benzene ring (Figure 4). This bicyclic heterocycle is capable of performing interactions with various biological targets (Ref Reference Carvalho, Luque-Ortega, López-Martín, Castanys, Rivas and Gamarro82) and show a vast range of biological properties like anticoagulant, anticancer, antiviral, antitrypanosomal, antioxidant and also anti-leishmanial activities. Their biological activity as antimicrobials makes them attractive candidates for the development of antibacterial and antiprotozoal medicines. Coumarin derivatives are known to interfere with mitochondrial function and lately, several coumarins have been developed with mitochondrial target function that needs to be tested as anti-leishmanial agents (Ref Reference Ortega-Forte, Rovira, Gandioso, Bonelli, Bosch, Ruiz and Marchán86). Coumarins have been used to treat prostate cancer, renal cell carcinoma, leukemia (Ref Reference Küpeli Akkol, Genç, Karpuz, Sobarzo-Sánchez and Capasso84) and used as anticoagulants (Ref Reference Flores-Morales, Villasana-Ruíz, Garza-Veloz, González-Delgado and Martinez-Fierro87).

Studies conducted in silico, in vitro and in vivo demonstrate that coumarins are suitable candidates for assessment as anti-leishmanial medications (Ref Reference Mandlik, Patil, Bopanna, Basu and Singh88). Five coumarins selected based on in silico studies were tested against CL. One derivative C2(6-Amino-3-(1,3-benzodioxol-5-yl)-2H-chromen-2-one) was used as nanoliposomal formulation against L. major promastigotes and significant anti-leishmanial activity was observed. There was a loss of ΔΨm after treatment with the compound in promastigotes. Continuing further with the study, testing in in vivo conditions showed a reduction in CL lesion sizes formed by L. major in BALB/c mice (Ref Reference Mandlik, Patil, Bopanna, Basu and Singh88). Mammea A/BB, a coumarin isolated from leaves of Calophyllum brasiliense showed anti-leishmanial activity on both promastigotes and amastigotes of L. amazonensis with mitochondrial effects. Good in vivo activity was visible in the reduced size of skin lesions in the footpads of BALB/c mice (Ref Reference Cardoso, De Mello, Lera, Brenzan, Cortez, Donatti and Lonardoni89, Reference Brenzan, Nakamura, Prado Dias Filho, Ueda-Nakamura, Young and Aparício Garcia Cortez90). Treatment with Mammea A/BB induced changes in the mitochondria of both promastigotes and amastigotes in terms of mitochondrial swelling with reduced density of the mitochondrial matrix and extra vesicles visible inside the mitochondrion, indicating damage and significant change in this organelle (Ref Reference Brenzan, Nakamura, Prado Dias Filho, Ueda-Nakamura, Young and Aparício Garcia Cortez90, Reference Tiuman, Brenzan, Ueda-Nakamura, Filho, Cortez and Nakamura91). The details of any effect on the ETC are not known. Another coumarin derivative, Mammea A/BA showed IC50 of 85.8 and 36.9 μM for epimastigotes and trypomastigotes, respectively, of Trypanosoma cruzi a related parasite where a significant increase in ROS with decrease ΔΨm was observed (Ref Reference Rodríguez-Hernández, Martínez, Reyes-Chilpa and Espinoza92). Subcutaneous treatment with (+)-3-(1′-dimethylallyl)-decursinol and (-)-heliettin, two coumarins extracted from Helietta apiculata Benth reduced size of cutaneous lesions generated in BALB/c mice with L. amazonensis by 95.6% and 98.6%, respectively (Ref Reference Ferreira, de Arias, Yaluff, de Bilbao, Nakayama, Torres and Fournet93). Osthole, a natural coumarin displayed dose-dependent leishmanicidal activity against intracellular amastigotes with IC50 value of 14.95 μg/ml. CL lesions generated in BALB/c mice with osthole resulted in declined lesion progression in comparison to suitable controls (Ref Reference Kermani, Sajjadi, Hejazi, Arjmand, Saberi and Eskandarian94). Earlier, aurapten, a 7-geranyloxy-coumarin was shown to have anti-leishmanial activity (Ref Reference Napolitano, Silva, Ellena, Rodrigues, Almeida, Vieira and Thiemann95). A review of natural and synthetic coumarins with anti-leishmanial efficacy details several important experiments and discusses the possibility of obtaining novel coumarin-based inhibitors (Ref Reference Gonçalves, Spillere, das Neves, Kagami, von Poser, Canto and Eifler-Lima96). To identify the ETC complex component that coumarins affect, further study is required but coumarins could be considered as potential anti-leishmanials.

Quinolones

Simple 3 carboxy quinolones were first described by J.R. Price and L.J. Drummond in 1949 (Ref Reference Bisacchi97). Quinoline alkaloids are secondary metabolites found in plants and are also produced by some bacterial species. Quinoline alkaloids have a wide range of biological activities including activity against cancer, malaria, inflammation and viruses (Ref Reference Yun, Yoon, Park and Park98, Reference Shang, Morris-Natschke, Liu, Guo, Xu, Goto and Lee99). Numerous quinolones like quinine, quinidine, cinchonine and cinchonidine have been obtained from natural sources since quinine was first extracted from the bark of the Cinchona plant in 1811 (Ref Reference Heeb, Fletcher, Chhabra, Diggle, Williams and Cámara100). Some of these have excellent medicinal properties and served as lead structures that provided the basis for synthetic quinolones as useful drugs (Ref Reference Heeb, Fletcher, Chhabra, Diggle, Williams and Cámara100). A few of these are very effective medications and were the model compounds from which synthetic quinolones were derived (Ref Reference Heeb, Fletcher, Chhabra, Diggle, Williams and Cámara100). Isolated from plant or microbial sources, several 4-quinolone alkaloids showed antimicrobial activities. The core structure is bicyclic (Figure 4; Ref Reference Pham, Ziora and Blaskovich101). Endochin-like quinolones (ELQs) show promise as novel antiparasitic drugs, they are analogs of ubiquinone that can act as competitive inhibitors of cytochrome bc1 by binding to the Qi ubiquinone binding site (Ref Reference McConnell, Bruzual, Pou, Winter, Dodean, Smilkstein and Doggett102, Reference Ortiz, Forquer, Boitz, Soysa, Elya, Fulwiler and Landfear103).

Studies show the efficacy of quinolones against CL caused by L. amazonensis and L. venezuelensis in infected BALB/c mice (Ref Reference Coimbra, Antinarelli, Silva, Souza, Meinel, Rocha and da Silva104). Other quinoline alkaloids isolated from the Galipea longiflora Krause D and B act against L. braziliensis and L. donovani promastigotes (Ref Reference Calla-Magariños, Quispe, Giménez, Freysdottir, Troye-Blomberg and Fernández105). N-methyl-8-methoxyflindersin, isolated from the leaves of Raputia heptaphylla also known as 7-methoxy-2,2-dimethyl-2H,5H,6H-pyran[3,2-c]quinolin-5-one, shows anti-parasitic activity against Leishmania promastigotes and amastigotes (Ref Reference Torres Suarez, Granados-Falla, Robledo, Murillo, Upegui and Delgado106). In silico tools used to identify synthetic quinoline alkaloids with similar structure to that of 7-methoxy-2,2-dimethyl-2H,5H,6H-pyran[3,2-c]quinolin-5-one tested for their in vitro antiparasitic activity against Leishmania (Viannia) panamensis, and in vivo therapeutic response in hamsters with experimental cutaneous leishmaniasis (CL) was reported (Ref Reference Torres Suarez, Granados-Falla, Robledo, Murillo, Upegui and Delgado106). Endochin like quinolones deplete ATP in promastigotes and amastigotes but at a slow pace (Ref Reference McConnell, Bruzual, Pou, Winter, Dodean, Smilkstein and Doggett102). Buparvaquone is a hydroxynaphthoquinone that is also related in structure to ubiquinone and is another potential inhibitor of cytochrome bc1 (Ref Reference McConnell, Bruzual, Pou, Winter, Dodean, Smilkstein and Doggett102). This compound has demonstrated efficacy against L. donovani in vitro and in a BALB/c model of visceral leishmaniasis (Ref Reference Coy Barrera, Coy Barrera, Granados Falla, Delgado Murcia and Cuca Suarez107). Buparvaquone and its phosphate prodrugs (BPQ-3-PHOS and 3-POM-BPQ) were tested against several species of Leishmania (L. major, L. amazonensis, L. aethiopica, L. mexicana and L. panamensis) in vitro. BPQ, BPQ-3-PHOS and 3-POM-BPQ demonstrated low IC50 values against promastigotes and amastigotes of all Leishmania species tested (Ref Reference Croft, Hogg, Gutteridge, Hudson and Randall108). Buparvaquone is a potent inhibitor of electron transport and robustly depletes ATP at submicromolar concentrations within an hour in vitro resulting in amastigote death. Buparvaquone’s toxicity may stem from its capacity to enhance reactive oxygen species (ROS) production, possibly as a result of its inhibition of cytochrome bc1 activity (Ref Reference Menna-Barreto and de Castro41). Buparvaquone significantly eliminates L. donovani parasites in mice models of leishmaniasis (Ref Reference Coy Barrera, Coy Barrera, Granados Falla, Delgado Murcia and Cuca Suarez107, Reference Luque-Ortega, Rivero-Lezcano, Croft and Rivas109). In an in vivo experiment, buparvaquone loaded with lipid nanoparticles with enhanced solubility demonstrates a 80% reduction in the livers’ burden of L. infatum (Ref Reference Smith, Serrano, Mauger, Bolás-Fernández, Dea-Ayuela and Lalatsa110, Reference Monteiro, Löbenberg, Barbosa, de Araujo, Sato, Kanashiro and Bou Chacra111). In a different study, lipid nanoparticles coated with buparvaquone demonstrated 100% clearance of parasites in an in vivo model, whereas pure buparvaquone demonstrated only 50% efficiency in this regard (Ref Reference Thapa, Mondal, Riikonen, Rantanen, Näkki, Nissinen and Lehto112). Cytochrome bc1 is thus a promising target for novel anti-leishmanial drugs, and further improvements on the buparvaquone scaffold are warranted for the development of enhanced therapeutics.

Quinolinyl β-enaminone derivatives showed anti-leishmanial activity through disruption of the mitochondrial ETC complex and induction of PCD after significant ROS production (Ref Reference Frosch, Schmitt, Bringmann, Kiefer and Popp66). Of the several compounds reported, the disruption of complex II by compound 3D ((Z)-1-(4-chlorophenyl)-3-(quinolin-8-ylamino)prop-2-en-1-one) was followed by oxidative burst, intracellular Ca2+ accumulation and activation of programmed cell death. In silico studies with compound 3D showed that mitochondrial complex II (SDH) or complex III (CCR) of L. donovani, modelled using the SWISS-Model server had more affinity towards SDH over CCR, which invariably correlates with its higher anti-leishmanial efficacy of 3D. This compound showed superior efficacy in vivo as well in infected BALB/c mice and hamsters (Ref Reference Rani, Khanikar, Dutta, Katiyar, Qamar, Seth and Kar70) indicating this as a potential compound that can be tested for therapeutic purposes.

Quercetin

Quercetin is a polyphenolic flavonoid with antioxidant characteristics found in a wide range of foods we consume, including citrus fruits and green leafy vegetables (Ref Reference Qi, Xiong and Long113). It was first isolated in 1854 (Ref Reference Ghareeb, Zayan, Shari, Sayed and Osredkar114). In essence, quercetin is a pentahydroxyflavone with five hydroxy groups positioned at positions 3, 3, 3’, 4, 5 and 7 (Figure 4) and is known to possess anti-inflammatory, anti-proliferative, anti-oxidative, anti-diabetic, anti-carcinogenic and anti-viral properties. Several in vivo and in vitro studies demonstrate the role of quercetin in various diseases and a detailed review is available (Ref Reference Deepika and Maurya115). The potential of quercetin as an anti-leishmanial agent was described in an early review (Ref Reference Sarkar, Mandal, Sinha, Mukhopadhyay, Das and Basu116). Following the release of the review, in vitro and in vivo investigations provided evidence supporting quercetin‘s potential as an anti-leishmanial agent. L. amazonensis and L. (V.) braziliensis, the causative agents of CL, L. donovani responsible for VL are sensitive to quercetin in vitro (Ref Reference Sousa-Batista, Poletto, Philipon, Guterres, Pohlmann and Bergmann117Reference Cataneo, Tomiotto-Pellissier, Miranda-Sapla, Assolini, Panis, Kian and Pavanelli119) and mitochondrial disruption is observed in both L. amazonensis and L. (V.) braziliensis along with ROS production (Ref Reference Cataneo, Tomiotto-Pellissier, Miranda-Sapla, Assolini, Panis, Kian and Pavanelli119, Reference Fonseca-Silva, Inacio, Canto-Cavalheiro and Almeida-Amaral120). In vivo studies in a murine model of CL created by L. amazonensis, a reduction in lesion size was observed (Ref Reference Muzitano, Falcão, Cruz, Bergonzi, Bilia, Vincieri and Costa121). Effective against both L. (V) brazilensis promastigotes and intracellular amastigotes, quercetin showed anti-amastigote IC50 (21 ± 2.5 μM) and anti-promastigote IC50 (25 ± 0.7 μM) activities with a selectivity index of 22 (Ref Reference Dos Santos, Da Silva, Brito, Inácio, Ventura, Mendes and Da Silva122). Quercetin was also effective as an oral drug in hamsters with CL (Ref Reference Dos Santos, Da Silva, Brito, Inácio, Ventura, Mendes and Da Silva122, Reference Ribeiro-Romão, Moreira, Osorio, Cysne-Finkelstein, Gomes-Silva, Valverde and Pinto123). Earlier studies with CL induced in BALB/c mice using L. amazonensis showed a reduction in lesion size with quercetin (Ref Reference Muzitano, Falcão, Cruz, Bergonzi, Bilia, Vincieri and Costa121). Five weeks after inoculation, CL generated in BALB/c mice using L. major showed a dramatic reduction in lesion size following oral quercetin exposure for a sequential 28 days, (Ref Reference Dos Santos, Da Silva, Brito, Inácio, Ventura, Mendes and Da Silva122). The anti-leishmanial impact of CL made with L. amazonensis in BALB/c mice was enhanced by the encapsulation of quercetin in lipid nanocapsules of poly(ϵ-caprolactone) (Ref Reference Sousa-Batista, Poletto, Philipon, Guterres, Pohlmann and Bergmann117). Quercetin may be taken into consideration for additional development given its effectiveness as an anti-leishmanial agent, as demonstrated by investigations both in vitro and in vivo.

Quercetin exerts its anti-leishmanial effect on L. amazonensis promastigotes through the disruption of mitochondria and generation of ROS (Ref Reference Fonseca-Silva, Inacio, Canto-Cavalheiro and Almeida-Amaral120). In mammalian cells, quercetin shows redox interaction with the mitochondrial ETC affecting its membrane potential and reducing ATP production. A review detailing interactions of quercetin with the ETC is available (Ref Reference Carrillo-Garmendia, Madrigal-Perez and Regalado-Gonzalez124). Studies with Leishmania mitochondria need to be explored further to assess the effect of quercetin on mitochondria.

In vitro tested plant compounds that affect the mitochondria

The following compounds have not been tested in higher animal models but are tested in vitro for efficacy with different Leishmania species and their effect on mitochondria has been analyzed. These could provide clues to taking up compounds for further studies for the development of anti-leishmanial agents. In L. amazonensis promastigotes, phytol-rich hexane fraction from the leaves of Lacistema pubescens induces depolarization of the mitochondria with an increase in ROS levels (Ref Reference da Silva, Antinarelli, Ribeiro, Coimbra and Scio125). Xanthohumol and resveratrol are naturally occurring constituents of the human diet. Resveratrol is formed as a phytoalexin in plants to defend against parasitic and fungal infections while xanthohumol belongs to the group of polyphenols. When tested with Leishmania parasites, both exerted anti-leishmanial action but in contrast to Resveratrol, xanthohumol strongly inhibited oxygen consumption in Leishmania tarantolae promastigotes. This was due to the inhibition of the mitochondrial electron transfer complex II/III by xanthohumol, which was less pronounced with Resveratrol (Ref Reference Monzote, Lackova, Staniek, Steinbauer, Pichler, Jäger and Gille126). Essential oil from Chenopodium ambrosioides L. and caryophyllene oxide, a compound derived from the essential oil was able to inhibit the electron transport chain of L. tarantolae promastigotes depriving the parasite of energy sources (Ref Reference Monzote, Lackova, Staniek, Steinbauer, Pichler, Jäger and Gille126). Iron can generate toxic ROS via the Fenton reaction. Studies to establish the influence of iron on the anti-leishmanial action of artemisinin in vitro showed that in Sneider’s insect medium, artemisinin caused a greater redox imbalance as compared to M199, a culture media where many experiments on Leishmania parasites grown in vitro are done (Ref Reference Dighal, De Sarkar, Gille and Chatterjee127). Therefore, the media type should be taken into account while analyzing the Leishmania in vitro tests. Several other compounds tested for anti-leishmanial effects through disruption of mitochondria primarily tested in vitro are described in a recent review (Ref Reference Lazarin-Bidóia, Garcia, Ueda-Nakamura, Silva and Nakamura128). Berberine chloride is an alkaloid that shows anti-leishmanial effects where death is preceded by an upsurge in the generation of ROS along with a depletion of ATP. In addition, mitochondrial superoxide, depolarization of the mitochondrial membrane potential, dose-dependent inhibition of mitochondrial complexes I-III and II-III collectively suggest interference with mitochondria in L. donovani (Ref Reference De Sarkar, Sarkar, Sarkar, Dighal, Staniek, Gille and Chatterjee129). Plumbagin, a naphthoquinone derivative results in the depolarization of the mitochondrial membrane, and depletion in ATP levels in the promastigotes of L. donovani inducing apoptosis-like death (Ref Reference Awasthi, Kathuria, Pant, Kumari and Mitra130). Essential oil from Chenopodium ambrosioides, an aromatic herb, kills the parasite by causing a breakdown of mitochondrial membrane potential and a modification of redox indexes (Ref Reference Zinovkin and Zamyatnin35). Formulations of saponins and hydrazones cause changes in the ATP levels and induce depolarization of membrane potential with ROS increase (Ref Reference Upegui Zapata, Echeverri, Quiñones, Torres, Nacher, Rivas and Robledo131). Ethanolic extract of Croton blanchetianus Ball is capable of causing depolarization of the mitochondria in Leishmania amazonensis promastigotes leading to death (Ref Reference Pereira, Vasconcelos, Braz, InÁcio, Estevam, Correa and Scher132). Phloroglucinol derivatives from Hypericum species induce mitochondrial dysfunction like hyperpolarization and ROS generation in promastigotes of L. amazonensis promastigotes (Ref Reference Dagnino, Mesquita, Dorneles, Teixeira, de Barros, Vidal Cana-Capatinta and Romão133). A naturally occurring plant flavone Apigenin, shows anti-leishmanicidal action against L. amazonensis promastigotes through induction of swelling of the parasite mitochondria resulting in a loss of mitochondrial membrane potential with accompanying ROS increase (Ref Reference Fonseca-Silva, Canto-Cavalheiro, Menna-Barreto and Almeida-Amaral134). Melatonin, a neurohormone found in animals, plants and microbes used in humans as medication changes calcium homeostasis of the mitochondrion of L. infantum, with the opening of the mitochondrial permeability transition pore, loss of ATP and mitochondrial potential loss (Ref Reference Elmahallawy, Jiménez-Aranda, Martínez, Rodriguez-Granger, Navarro-Alarcón, Gutiérrez-Fernández and Agil135). Two plant-based compounds, piperine and phenylamide eliminate Leishmania promastigotes and amastigotes through induction of mitochondrial swelling and a loss of mitochondrial membrane potential (Ref Reference Ferreira, Soares, Barreto-Junior, Nascimento, Freire-de-Lima, Delorenzi and Pinto-da-Silva69). Caffeic acid shows anti-leishmanial activity against promastigotes and amastigotes of Leishmania amazonensis by the increase of oxidant activities (Ref Reference Bortoleti, Tomiotto-Pellissier, Gonçalves, Miranda-Sapla, Assolini, Carloto and Pavanelli136). In mammalian cancer cells a variety of caffeic acid derivatives targeted to mitochondria have been developed (Ref Reference Bastidas, Araya-Valdés, Cortés, Jara and Catalán36). These should be tested in the Leishmania parasites both in vitro and in vivo. Curcumin (1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) is a natural polyphenol found in the rhizomes of Curcuma spp. and is a well-known diferuloylmethane. Curcumin is a potent antioxidant agent with anti-inflammatory and anti-tumor properties. In several species of Leishmania, curcumin causes mitochondrial membrane depolarization along with hyperproduction of ROS (Ref Reference Das, Roy, Dutta and Majumder137). In cancer cells, curcumin was shown to cause mitochondrial disruption leading to ROS production (Ref Reference Jung, Lee, Park, Moon, Cho, Choe and Lee138).

Discussion

The growing global usage of herbal products raises the possibility that plant-based compounds could be a rich source of anti-leishmanials (Ref Reference Chaughule and Barve139). For any potential compound for use to counter pathogens, it is important to identify a susceptible cellular target in the pathogen, accessible to the drug. Mitochondria are one such organelle that are appropriate targets of multiple drugs (Ref Reference Zinovkin and Zamyatnin35) because functionally they maintain cellular homeostasis through energy production and any interference with their operation is detrimental to the cells. The single mitochondrion of the Leishmania parasite is a prime target as its functional failure would jeopardize the parasite’s survival ability. In the last decade, many mitochondria-targeted drugs have been synthesized and tested in the mammalian system and some are in clinical trials (Ref Reference Zinovkin and Zamyatnin35). Currently, multiple mitochondria-targeted drugs are being tested in mammalian cancer cells (Ref Reference Bastidas, Araya-Valdés, Cortés, Jara and Catalán36, Reference Dong, Zheng, Gao, Li and Zhong140) and some of these could be useful as new drugs for leishmaniasis.

Artemisinin and its derivatives have enormous therapeutic value as antimalarials (Ref Reference Guan, Wang, Xu and Fan141) and have been repurposed for viral infections, autoimmune conditions and cancer (Ref Reference Chaughule and Barve139). Such efforts indicate the possibility of artemisinin being repurposed as anti-leishmanials because of their efficacy in both models of VL and CL. This has been substantiated by in vitro and in vivo investigations (Ref Reference Dong, Liu, Han, Zhang, Wang, Li and Hu142). Additionally, artemisinin and its derivatives target the mitochondrion of the Leishmania making them effective for both forms of the parasite. Chloroquine and its derivatives are used to treat a variety of illnesses, with its efficacy as an anti-leishmanial shown in in vivo testing (Ref Reference Kamat and Kumari143). Coumarins have been thoroughly investigated as medications for different diseases (Ref Reference Flores-Morales, Villasana-Ruíz, Garza-Veloz, González-Delgado and Martinez-Fierro87) and are good candidates for anti-leishmanial treatment, especially because they show in vivo efficacy against the Leishmania parasite. Quinolones are considered to have privileged structures that can be manipulated to generate novel molecules. Quinoline alkaloids show significant anti-leishmanial activities both in vitro and in vivo and therefore with their background of being tested in humans are good candidates for development as anti-leishmanial drugs. Reported to affect the Leishmania mitochondria (Ref Reference Fonseca-Silva, Inacio, Canto-Cavalheiro and Almeida-Amaral120) quercetin is a good candidate to be considered as a leishmanicidal drug. A summary of the compounds analyzed are detailed in Table 1. In summary, the promising compounds detailed here should be taken up for further investigations for use as anti-leishmanials, as their use has already been tested in the human system.

Table 1 Table describing the plants from which the compounds were first prepared, the part of the plants from where the extracts were made, and the targets of the compounds on the parasite mitochondrion

Future directions

The plethora of plant-based compounds and their derivatives with evidence of leishmanicidal activities, offer new opportunities for the field of anti-leishmanials. The possible drugs analyzed above should be taken forward by filling the gaps necessary to satisfy preclinical requirements. Taking lessons from the field of cancer drug development, mitochondria-targeted drugs now being designed for cancer and other diseases (Ref Reference Dhanasekaran, Venugopal, Al-Dayan, Ravinayagam and Mohammed37, Reference Jeena, Kim, Jin and Ryu144) could form a repertoire of possible new anti-leishmanial compounds. Some derivatives of artemisinin synthesized specifically targeting the mitochondria in mammalian cells (Ref Reference Xu, Xiao, Zhang, Zhuang, Mu and Yang145) have not been tested in the Leishmania parasite. These compounds with low toxicity in in vitro studies with mammalian cells indicate the suitability of their use as potential agents suitable for pathogen elimination (Ref Reference Xu, Xiao, Zhang, Zhuang, Mu and Yang145). The chloroquine derivative DNDI-0690 has completed clinical trials for leishmaniasis, further designing of effective drugs based on the structure of this compound has the possibility of obtaining more effective anti-leishmanials. With the high efficacy of the 4-aminoquinoline derivatives against leishmaniasis, further studies are required to generate alternatives to DNDI-0690.

It is expected that researchers will benefit from this review built to detail compounds with in vivo efficacy and with effects on the mitochondria. Over the past several years, a significant effort has gone into in silico studies where molecular docking has been used to identify potential compounds for anti-leishmanial efficacy, even predicting the structure of their binding sites (Ref Reference Scotti, Ishiki, Mendonça Júnior, Da Silva and Scotti146Reference Kant, Kumar, Ranjan, Mandal and Vijayakumar149).

A strategy for discovering new uses for approved drugs under investigation for repurposing could be outside the scope of the original medication. One of the major benefits of repurposing is that the drugs have already been tested in humans and have on record full information on their pharmacology, dose, possible toxicity and formulation. This cuts the cost of research and development, it does not require phase I clinical trials in humans and toxicity data are available. Artemisinin, chloroquine and their derivatives known for their efficacy in malaria have been tested in animal studies for leishmaniasis with success. Therefore, the future of drug development against leishmaniasis could firmly advance the findings over the past years on the in vivo efficacy with improved drug delivery systems. Other suggestions have been made, for example, the use of structural biology to look for drug targets for leishmaniasis (Ref Reference Khandibharad and Singh150) and, the use of molecular docking and molecular dynamic simulations for anti-leishmanial drug development (Ref Reference Challapa-Mamani, Tomás-Alvarado, Espinoza-Baigorria, León-Figueroa, Sah, Rodriguez-Morales and Barboza151). Lessons from different fields of research if applied judiciously would provide a huge source of possible plant-based drugs for several diseases. Joint collaborative research in interdisciplinary areas is required to focus on relevant structures of compounds that offer prospects for drug development. Given the complexity of drug development, multiple stakeholders need to be involved. For efficient bench-to-bedside translation, cooperation between the industry and academia is a must as knowledge generation and making formulations suitable for patient use have to go hand in hand. The medical fraternity here plays an important role as they have a complete understanding of diseases. For them to be in the loop not only during preclinical trials but also during drug development is a necessity.

Acknowledgements

The study was supported by funding from the National Academy of Sciences, Allahabad under the J.C. Bose Chair Distinguished Professor Scheme (NASI/174/7/2021) and the Indian National Science Academy (No. 1353r2), New Delhi. Facilities were provided by the Indian Institute of Chemical Biology, Kolkata.

References

Mathur, S and Hoskins, C (2017) Drug development: Lessons from nature. Biomedical Reports 6(6), 612614. https://doi.org/10.3892/br.2017.909Google Scholar
Christensen, SB (2021) Natural products that changed society. Biomedicines 9(5), 472. https://doi.org/10.3390/biomedicines9050472Google Scholar
Theodoridis, S, Drakou, EG, Hickler, T, Thines, M and Nogues-Bravo, D (2023) Evaluating natural medicinal resources and their exposure to global change. Lancet Planet Health 7(2), e155e163. https://doi.org/10.1016/S2542-5196(22)00317-5Google Scholar
Alcazar, W, Alakurtti, S, Padrón-Nieves, M, Tuononen, ML, Rodríguez, N, Yli-Kauhaluoma, J and Ponte-Sucre, A (2021) Leishmanicidal activity of betulin derivatives. Microorganisms 9(2), 320. https://doi.org/10.3390/microorganisms9020320Google Scholar
Atanasov, AG, Zotchev, SB, Dirsch, VM, Supuran, CT and Taskforce, INPS (2021) Natural products in drug discovery: Advances and opportunities. Nature Reviews Drug Discovery 20(3), 200216. https://doi.org/10.1038/s41573-020-00114-zGoogle Scholar
Ghorbani, M and Farhoudi, R (2018) Leishmaniasis in humans: Drug or vaccine therapy? Drug Design, Development and Therapy 12 2540.Google Scholar
Charlton, RL, Rossi-Bergmann, B, Denny, PW and Steel, PG (2018) Repurposing as a strategy for the discovery of new anti-leishmanials: The-state-of-the-art. Parasitology 145(2), 219236. https://doi.org/10.1017/S0031182017000993Google Scholar
Bustamante, C, Ochoa, R, Asela, C and Muskus, C (2019) Repurposing of known drugs for leishmaniasis treatment using bioinformatic predictions, in vitro validations and pharmacokinetic simulations. Journal of Computer-Aided Molecular Design 33(9), 845854. https://doi.org/10.1007/s10822-019-00230yGoogle Scholar
Akhoundi, M, Downing, T, Votýpka, J, Kuhls, K, Lukeš, J, Cannet, A,…Sereno, D (2017) Leishmania infections: Molecular targets and diagnosis. Molecular Aspects of Medicine 57, 129. https://doi.org/10.1016/j.mam.2016.11.012Google Scholar
Hotez, PJ (2018) The rise of leishmaniasis in the twenty-first century. Transactions of the Royal Society of Tropical Medicine and Hygiene 112(9), 421422. https://doi.org/10.1093/trstmh/try075Google Scholar
Boodman, C, van Griensven, J, Gupta, N, Diro, E and Ritmeijer, K (2023) Anticipating visceral leishmaniasis epidemics due to the conflict in Northern Ethiopia. PLOS Neglected Tropical Diseases 17(3), e0011188. https://doi.org/10.1371/journal.pntd.0011188Google Scholar
Al-Salem, WS, Pigott, DM, Subramaniam, K, Haines, LR, Kelly-Hope, L, Molyneux, DH,…Acosta-Serrano, A (2016) Cutaneous leishmaniasis and conflict in Syria. Emerging Infectious Diseases 22(5), 931933. https://doi.org/10.3201/eid2205.160042Google Scholar
Torres-Guerrero, E, Quintanilla-Cedillo, MR, Ruiz-Esmenjaud, J and Arenas, R (2017) Leishmaniasis: A review. F1000Research 6, 750. https://doi.org/10.12688/f1000research.11120.1Google Scholar
Kumar, A, Singh, VK, Tiwari, R, Madhukar, P, Rajneesh, Kumar S,…Kumar, R (2023) Post kala-azar dermal leishmaniasis in the Indian sub-continent: Challenges and strategies for elimination. Frontiers in Immunology 14, 1236952. https://doi.org/10.3389/fimmu.2023.1236952Google Scholar
Kevric, I, Cappel, MA and Keeling, JH (2015) New world and old world Leishmania infections: A practical review. Dermatologic Clinics 33(3), 579593. https://doi.org/10.1016/j.det.2015.03.018Google Scholar
Miret, JA, Moreno, J, Nieto, J, Carter, KC, Mullen, AB, Ambros, L,…González, F (2021) Anti-leishmanial efficacy and tolerability of combined treatment with non-ionic surfactant vesicle formulations of sodium stibogluconate and paromomycin in dogs. Experimental Parasitology 220, 108033. https://doi.org/10.1016/j.exppara.2020.108033Google Scholar
Olías-Molero, AI, de la Fuente, C, Cuquerella, M, Torrado, JJ and Alunda, JM (2021) Anti-leishmanial drug discovery and development: Time to reset the model? Microorganisms 9(12), 2500. https://doi.org/10.3390/microorganisms9122500Google Scholar
Sundar, S and Olliaro, PL (2007) Miltefosine in the treatment of leishmaniasis: Clinical evidence for informed clinical risk management. Therapeutics and Clinical Risk Management 3(5), 733740.Google Scholar
Jhingran, A, Chawla, B, Saxena, S, Barrett, MP and Madhubala, R (2009) Paromomycin: uptake and resistance in Leishmania donovani. Molecular and Biochemical Parasitology 164(2), 111117. https://doi.org/10.1016/j.molbiopara.2008.12.007Google Scholar
Clos, J, Grünebast, J and Holm, M (2022) Promastigote-to-Amastigote Conversion in Leishmania spp.-A molecular view. Pathogens 11(9), 1052 https://doi.org/10.3390/pathogens11091052Google Scholar
Paun, A (2022) Culture of cutaneous Leishmania from skin biopsy specimens. Current Protocols 2(2), e367. https://doi.org/10.1002/cpz1.367Google Scholar
Sundar, S and Rai, M (2002) Laboratory diagnosis of visceral leishmaniasis. Clinical and Diagnostic Laboratory Immunology 9(5), 951958. https://doi.org/10.1128/cdli.9.5.951-958.2002Google Scholar
Arjmand, M, Madrakian, A, Khalili, G, Najafi Dastnaee, A, Zamani, Z and Akbari, Z (2016) Metabolomics-based study of logarithmic and stationary phases of promastigotes in Leishmania major by 1H NMR spectroscopy. Iranian Biomedical Journal 20(2), 7783. https://doi.org/10.7508/ibj.2016.02.002Google Scholar
Sudhandiran, G and Shaha, C (2003) Antimonial-induced increase in intracellular Ca2+ through non-selective cation channels in the host and the parasite is responsible for apoptosis of intracellular Leishmania donovani amastigotes. Journal of Biological Chemistry 278(27), 2512025132. https://doi.org/10.1074/jbc.M301975200Google Scholar
Chanmol, W, Jariyapan, N, Somboon, P, Bates, MD and Bates, PA (2019) Axenic amastigote cultivation and in vitro development of Leishmania orientalis. Parasitology Research 118(6), 18851897. https://doi.org/10.1007/s00436-019-06311-zGoogle Scholar
Loría-Cervera, EN and Andrade-Narváez, FJ (2014) Animal models for the study of Leishmaniasis immunology. Revista do Instituto de Medicina Tropical de São Paulo 56(1), 111. https://doi.org/10.1590/S0036-46652014000100001Google Scholar
Shivahare, R, Vishwakarma, P, Parmar, N, Yadav, PK, Haq, W, Srivastava, M,…Kar, S (2014) Combination of liposomal CpG oligodeoxynucleotide 2006 and miltefosine induces strong cell-mediated immunity during experimental visceral leishmaniasis. PLoS One 9(4), e94596. https://doi.org/10.1371/journal.pone.0094596Google Scholar
Kathuria, M, Bhattacharjee, A, Sashidhara, KV, Singh, SP, Mitra, K (2014) Induction of mitochondrial dysfunction and oxidative stress in Leishmania donovani by orally active clerodane diterpene. Antimicrobial Agents and Chemotherapy 58(10), 59165928. https://doi.org/10.1128/AAC.02459-14Google Scholar
Camacho, E, Rastrojo, A, Sanchiz, Á, González-de la Fuente, S, Aguado, B, Requena, JM (2019) Mitochondrial genomes: Maxicircle structure and heterogeneity of minicircles. Genes 10(10) https://doi.org/10.3390/genes10100758.Google Scholar
Pusnik, M, Mani, J, Schmidt, O, et al (2012) An essential novel component of the noncanonical mitochondrial outer membrane protein import system of trypanosomatids. Molecular Biology of the Cell 23(17):34203428. https://doi.org/10.1091/mbc.E12-02-0107.Google Scholar
Mehta, A and Shaha, C (2004) Apoptotic death in Leishmania donovani promastigotes in response to respiratory chain inhibition: Complex II inhibition results in increased pentamidine cytotoxicity. Journal of Biological Chemistry 279(12), 1179811813. https://doi.org/10.1074/jbc.M309341200Google Scholar
Mehta, A and Shaha, C (2006) Mechanism of metalloid-induced death in Leishmania spp.: role of iron, reactive oxygen species, Ca2+, and glutathione. Free Radical Biology and Medicine 40(10), 18571868. https://doi.org/10.1016/j.freeradbiomed.2006.01.024Google Scholar
Shadab, M, Jha, B, Asad, M, Deepthi, M, Kamran, M and Ali, N (2017) Apoptosis-like cell death in Leishmania donovani treated with KalsomeTM10, a new liposomal amphotericin B. PLoS One 12(2), e0171306. https://doi.org/10.1371/journal.pone.0171306Google Scholar
Zinovkin, RA and Zamyatnin, AA (2019) Mitochondria-targeted Drugs. Current Molecular Pharmacology 12(3), 202214. https://doi.org/10.2174/1874467212666181127151059Google Scholar
Bastidas, H, Araya-Valdés, G, Cortés, G, Jara, JA and Catalán, M (2022) Pharmacological effects of caffeic acid and its derivatives in cancer: New targeted compounds for the mitochondria. Advances in Experimental Medicine and Biology 1401, 213225. https://doi.org/10.1007/5584_2022_718Google Scholar
Dhanasekaran, S, Venugopal, D, Al-Dayan, N, Ravinayagam, V and Mohammed, AA (2020) Emerging insights into mitochondria-specific targeting and drug delivering strategies: Recent milestones and therapeutic implications. Saudi Journal of Biological Sciences 27(12), 35813592. https://doi.org/10.1016/j.sjbs.2020.07.030Google Scholar
Monzote, L and Gille, L (2010) Mitochondria as a promising antiparasitic target. Current Clinical Pharmacology 5(1), 5560. https://doi.org/10.2174/157488410790410605Google Scholar
Zorova, LD, Popkov, VA, Plotnikov, EY, Silachev, DN, Pevzner, IB, Jankauskas, SS,…Zorov, DB (2018) Mitochondrial membrane potential. Analytical Biochemistry 552, 5059. https://doi.org/10.1016/j.ab.2017.07.009Google Scholar
Ly, JD, Grubb, DR and Lawen, A (2003) The mitochondrial membrane potential (deltapsi(m)) in apoptosis; an update. Apoptosis 8(2), 115128. https://doi.org/10.1023/a:1022945107762Google Scholar
Menna-Barreto, RF and de Castro, SL (2014). The double-edged sword in pathogenic trypanosomatids: The pivotal role of mitochondria in oxidative stress and bioenergetics. BioMed Research Internationa 2014, 614014. https://doi.org/10.1155/2014/614014Google Scholar
Das, S Giri, S, Sundar, S, Shaha, C (2018) Functional involvement of Leishmania donovani tryparedoxin peroxidases during infection and drug treatment. Antimicrobial Agents and Chemotherapy 62(1), e00806e00817. https://doi.org/10.1128/AAC.00806-17.Google Scholar
Wang, J., Xu, C., Wong, YK, Liao, FL, Jiang, T and Tu, Y (2020) Malaria eradication. Lancet 395(10233), e69. https://doi.org/10.1016/S0140-6736(20)30223-3Google Scholar
Kong, LY and Tan, RX (2015) Artemisinin, a miracle of traditional Chinese medicine. Natural Product Reports 32(12), 16171621. https://doi.org/10.1039/c5np00133aGoogle Scholar
Chen, I and Hsiang, MS (2022). Triple artemisinin-based combination therapies for malaria: A timely solution to counter antimalarial drug resistance. The Lancet Infectious Diseases 22(6), 751753. https://doi.org/10.1016/S1473-3099(21)00748-9Google Scholar
Krishna, S, Bustamante, L, Haynes, RK and Staines, HM (2008) Artemisinins: Their growing importance in medicine. Trends in Pharmacological Sciences 29(10), 520527. https://doi.org/10.1016/j.tips.2008.07.004Google Scholar
Issa, MS, Warsame, M, Mahamat, MHT, Saleh, IDM, Boulotigam, K, Djimrassengar, H,…Menard, D (2023). Therapeutic efficacy of artesunate-amodiaquine and artemether-lumefantrine for the treatment of uncomplicated falciparum malaria in Chad: Clinical and genetic surveillance. Malaria Journal 22(1), 240. https://doi.org/10.1186/s12936-023-04644-wGoogle Scholar
Salas-Coronas, J, Vázquez-Villegas, J, Villarejo-Ordóñez, A, Sánchez-Sánchez, JC, Espada-Chavarría, J, Soriano-Pérez, MJ,…Cabezas-Fernández, MT (2013) [Radiological findings in patients with imported schistosomiasis]. Enfermedades Infecciosas y Microbiología Clínica 31(4), 205209. https://doi.org/10.1016/j.eimc.2012.04.003Google Scholar
Lam, NS, Long, X, Su, XZ and Lu, F (2018) Artemisinin and its derivatives in treating helminthic infections beyond schistosomiasis. Pharmacological Research 133(7), 77100. https://doi.org/10.1016/j.phrs.2018.04.025Google Scholar
Bergquist, R and Elmorshedy, H (2018) Artemether and praziquantel: origin, mode of action, impact, and suggested application for effective control of human schistosomiasis. Tropical Medicine and Infectious Disease 3(4), 125137. https://doi.org/10.3390/tropicalmed3040125Google Scholar
Sen, R, Bandyopadhyay, S, Dutta, A, Mandal, G, Ganguly, S, Saha, P and Chatterjee, M (2007) Artemisinin triggers induction of cell-cycle arrest and apoptosis in Leishmania donovani promastigotes. Journal of Medical Microbiology 56(Pt 9), 12131218. https://doi.org/10.1099/jmm.0.47364-0Google Scholar
Islamuddin, M, Chouhan, G, Tyagi, M, Abdin, MZ, Sahal, D and Afrin, F (2014) Leishmanicidal activities of Artemisia annua leaf essential oil against visceral leishmaniasis. Frontiers in Microbiology 5, 626. https://doi.org/10.3389/fmicb.2014.00626Google Scholar
Want, MY, Islammudin, M, Chouhan, G, Ozbak, HA, Hemeg, HA, Chattopadhyay, AP and Afrin, F (2017) Nanoliposomal artemisinin for the treatment of murine visceral leishmaniasis. International Journal of Nanomedicine 12, 21892204. https://doi.org/10.2147/IJN.S106548Google Scholar
Aderibigbe, BA (2017) Metal-based nanoparticles for the treatment of infectious diseases. Molecules 22(8) 1370. https://doi.org/10.3390/molecules22081370Google Scholar
Li, GQ, Guo, XB, Fu, LC, Jian, HX and Wang, XH (1994) Clinical trials of artemisinin and its derivatives in the treatment of malaria in China. Transactions of the Royal Society of Tropical Medicine and Hygiene 88 Suppl 1, S5S6. https://doi.org/10.1016/0035-9203(94)90460-xGoogle Scholar
Machín, L, Nápoles, R, Gille, L and Monzote, L (2021) Leishmania amazonensis response to artemisinin and derivatives. Parasitology International 80, 102218. https://doi.org/10.1016/j.parint.2020.102218Google Scholar
Gugliandolo, E, Palma, E, Peritore, AF, Siracusa, R, D’Amico, R, Fusco, R,…Crupi, R (2020) Effect of artesunate on Leishmania amazonesis induced neuroinflammation and nociceptive behavior in male balb/c mice. Animals (Basel) 10(4), 557. https://doi.org/10.3390/ani10040557Google Scholar
Ghaffarifar, F, Esavand Heydari, F, Dalimi, A, Hassan, ZM, Delavari, M and Mikaeiloo, H (2015) Evaluation of apoptotic and anti-Leishmanial activities of artemisinin on promastigotes and balb/c mice infected with Leishmania major. Iranian Journal of Parasitology 10(2), 258267.Google Scholar
De Sarkar, S, Sarkar, D, Sarkar, A, Dighal, A, Chakrabarti, S, Staniek, K,…Chatterjee, M (2019) The leishmanicidal activity of artemisinin is mediated by cleavage of the endoperoxide bridge and mitochondrial dysfunction. Parasitology 146 (4), 511520. https://doi.org/10.1017/S003118201800183XGoogle Scholar
Wang, J, Huang, L, Li, J, Fan, Q, Long, Y, Li, Y and Zhou, B (2010) Artemisinin directly targets malarial mitochondria through its specific mitochondrial activation. PLoS One 5(3), e9582. https://doi.org/10.1371/journal.pone.0009582Google Scholar
Geroldinger, G, Tonner, M, Hettegger, H, Bacher, M, Monzote, L, Walter, M,…Gille, L (2017) Mechanism of ascaridole activation in Leishmania. Biochemical Pharmacology 132, 4862. https://doi.org/10.1016/j.bcp.2017.02.023Google Scholar
Staniek, K, Gille, L, Kozlov, AV and Nohl, H (2002) Mitochondrial superoxide radical formation is controlled by electron bifurcation to the high and low potential pathways. Free Radical Research 36(4), 381387. https://doi.org/10.1080/10715760290021225Google Scholar
Antoine, T, Fisher, N, Amewu, R, O’Neill, PM, Ward, SA and Biagini, GA (2014) Rapid kill of malaria parasites by artemisinin and semi-synthetic endoperoxides involves ROS-dependent depolarization of the membrane potential. Journal of Antimicrobial Chemotherapy 69(4), 10051016. https://doi.org/10.1093/jac/dkt486Google Scholar
Grazzia, N, Boaventura, S, Garcia, VL, Gadelha, FR and Miguel, DC (2021). Dihydroartemisinin, an active metabolite of artemisinin, interferes with Leishmania braziliensis mitochondrial bioenergetics and survival. Parasitology Research 120(2), 705713. https://doi.org/10.1007/s00436-020-07019-1Google Scholar
Zhou, B and Yue, JM (2022) Natural products are the treasure pool for antimalarial agents. National Science Review 9(11), nwac112. https://doi.org/10.1093/nsr/nwac112Google Scholar
Frosch, T, Schmitt, M, Bringmann, G, Kiefer, W and Popp, J (2007) Structural analysis of the anti-malaria active agent chloroquine under physiological conditions. The Journal of Physical Chemistry B 111(7), 18151822. https://doi.org/10.1021/jp065136jGoogle Scholar
Carmo, AM., Silva, FM., Machado, PA, Fontes, AP, Pavan, FR, Leite, CQ,…Da Silva, AD (2011) Synthesis of 4-aminoquinoline analogues and their platinum(II) complexes as new anti-Leishmanial and antitubercular agents. Biomedicine & Pharmacotherapy 65(3), 204209. https://doi.org/10.1016/j.biopha.2011.01.003Google Scholar
Pandey, AK, Sharma, S, Pandey, M, Alam, MM, Shaquiquzzaman, M and Akhter, M (2016) 4, 5-Dihydrooxazole-pyrazoline hybrids: Synthesis and their evaluation as potential antimalarial agents. European Journal of Medicinal Chemistry 123, 476486. https://doi.org/10.1016/j.ejmech.2016.07.055Google Scholar
Ferreira, C, Soares, DC, Barreto-Junior, CB, Nascimento, MT, Freire-de-Lima, L, Delorenzi, JC,…Pinto-da-Silva, LH (2011) Leishmanicidal effects of piperine, its derivatives, and analogues on Leishmania amazonensis. Phytochemistry 72(17), 21552164. https://doi.org/10.1016/j.phytochem.2011.08.006Google Scholar
Rani, A, Khanikar, S, Dutta, M, Katiyar, S, Qamar, T, Seth, A,…Kar, S (2022) Quinolinyl β-enaminone derivatives exhibit leishmanicidal activity against Leishmania donovani by impairing the mitochondrial electron transport chain complex and inducing ROS-mediated programmed cell death. Journal of Antimicrobial Chemotherapy. 78, 359372. https://doi.org/10.1093/jac/dkac395Google Scholar
Singh, R, Kashif, M, Srivastava, P and Manna, PP (2023) Recent advances in chemotherapeutics for leishmaniasis: Importance of the cellular biochemistry of the parasite and its molecular interaction with the host. Pathogens 12(5), 706742. https://doi.org/10.3390/pathogens12050706Google Scholar
Verma, A and Saraf, SK ( 2008 ) 4-thiazolidinone–a biologically active scaffold. European Journal of Medicinal Chemistry 43(5), 897905.doi: 10.1016/j.ejmech.2007.07.017Google Scholar
Konstantinović, J, Videnović, M, Orsini, S, Bogojević, K, D’Alessandro, S, Scaccabarozzi, D,…Šolaja, BA (2018) Novel aminoquinoline derivatives significantly reduce parasite load in Leishmania infantum infected mice. ACS Medicinal Chemistry Letters 9(7), 629634. https://doi.org/10.1021/acsmedchemlett.8b00053Google Scholar
Loiseau, PM, Cojean, S and Schrével, J (2011) Sitamaquine as a putative anti-leishmanial drug candidate: From the mechanism of action to the risk of drug resistance. Parasite 18(2), 115119. https://doi.org/10.1051/parasite/2011182115Google Scholar
Jha, TK, Sundar, S, Thakur, CP, Felton, JM, Sabin, AJ and Horton, J (2005) A phase II dose-ranging study of sitamaquine for the treatment of visceral leishmaniasis in India. American Journal of Tropical Medicine and Hygiene 73(6), 10051011.Google Scholar
Gupta, S, Yardley, V, Vishwakarma, P, Shivahare, R, Sharma, B, Launay, D,…Puri, SK (2015) Nitroimidazo-oxazole compound DNDI-VL-2098: an orally effective preclinical drug candidate for the treatment of visceral leishmaniasis. Journal of Antimicrobial Chemotherapy 70(2), 518527. https://doi.org/10.1093/jac/dku422Google Scholar
Van Bocxlaer, K, McArthur, KN, Harris, A, Alavijeh, M, Braillard, S, Mowbray, CE and Croft, SL (2021) Film-forming systems for the delivery of DNDI-0690 to treat cutaneous Leishmaniasis. Pharmaceutics 13(4). https://doi.org/10.3390/pharmaceutics13040516Google Scholar
Wijnant, GJ, Croft, SL, de la Flor, R, Alavijeh, M, Yardley, V, Braillard, S,…Van Bocxlaer, K (2019) Pharmacokinetics and pharmacodynamics of the nitroimidazole DNDI-0690 in mouse models of cutaneous leishmaniasis. Antimicrobial Agents and Chemotherapy 63(9). https://doi.org/10.1128/AAC.00829-19Google Scholar
Antinarelli, LM, Dias, RM, Souza, IO, Lima, WP, Gameiro, J, da Silva, AD and Coimbra, ES (2015) 4-Aminoquinoline derivatives as potential anti-leishmanial agents. Chemical Biology & Drug Design 86(4), 704714. https://doi.org/10.1111/cbdd.12540Google Scholar
Herrera, L, Llanes, A, Álvarez, J, Degracia, K, Restrepo, CM, Rivera, R,…Fernández, PL (2020) Anti-leishmanial activity of a new chloroquine analog in an animal model of Leishmania panamensis infection. International Journal for Parasitology: Drugs and Drug Resistance 14, 5661. https://doi.org/10.1016/j.ijpddr.2020.08.002Google Scholar
Carvalho, L., Luque-Ortega, JR, Manzano, JI, Castanys, S, Rivas, L and Gamarro, F (2010) Tafenoquine, an antiplasmodial 8-aminoquinoline, targets Leishmania respiratory complex III and induces apoptosis. Antimicrobial Agents and Chemotherapy 54 (12), 53445351. https://doi.org/10.1128/AAC.00790-10Google Scholar
Carvalho, L, Luque-Ortega, JR, López-Martín, C, Castanys, S, Rivas, L and Gamarro, F (2011) The 8-aminoquinoline analogue sitamaquine causes oxidative stress in Leishmania donovani promastigotes by targeting succinate dehydrogenase. Antimicrobial Agents and Chemotherapy, 55 (9), 42044210. https://doi.org/10.1128/AAC.00520-11Google Scholar
Wang, Z, Sheaff, RJ and Hussaini, SR (2023) Chloroquine-based mitochondrial ATP inhibitors. Molecules 28(3) 1161. https://doi.org/10.3390/molecules28031161Google Scholar
Küpeli Akkol, E, Genç, Y, Karpuz, B, Sobarzo-Sánchez, E and Capasso, R (2020) Coumarins and coumarin-related compounds in pharmacotherapy of cancer. Cancers (Basel) 12(7), 1959. https://doi.org/10.3390/cancers12071959Google Scholar
Stefanachi, A, Leonetti, F, Pisani, L, Catto, M and Carotti, A (2018) Coumarin: A natural, privileged and versatile scaffold for bioactive compounds. Molecules 23(2), 250. https://doi.org/10.3390/molecules23020250Google Scholar
Ortega-Forte, E, Rovira, A, Gandioso, A, Bonelli, J, Bosch, M, Ruiz, J and Marchán, V (2021) COUPY coumarins as novel mitochondria-targeted photodynamic therapy anticancer agents. J Med Chem 64(23), 1720917220. https://doi.org/10.1021/acs.jmedchem.1c01254Google Scholar
Flores-Morales, V, Villasana-Ruíz, AP, Garza-Veloz, I, González-Delgado, S and Martinez-Fierro, ML (2023) Therapeutic effects of coumarins with different substitution patterns. Molecules 28(5), 2413. https://doi.org/10.3390/molecules28052413Google Scholar
Mandlik, V, Patil, S, Bopanna, R, Basu, S and Singh, S (2016) Biological activity of coumarin derivatives as anti-leishmanial agents. PLoS One 11(10), e0164585. https://doi.org/10.1371/journal.pone.0164585Google Scholar
Cardoso, BM., De Mello, TF., Lera, DS., Brenzan, MA., Cortez, DA., Donatti, L.,…Lonardoni, MV (2017) Antileishmanial activity of a calophyllum brasiliense leaf extract. Planta Medica 83(1–02), 5762. https://doi.org/10.1055/s-0042-107673Google Scholar
Brenzan, MA, Nakamura, CV, Prado Dias Filho, B, Ueda-Nakamura, T, Young, MC and Aparício Garcia Cortez, D (2007) Antileishmanial activity of crude extract and coumarin from Calophyllum brasiliense leaves against Leishmania amazonensis. Parasitology Research 101(3), 715722. https://doi.org/10.1007/s00436-007-0542-7Google Scholar
Tiuman, TS, Brenzan, MA, Ueda-Nakamura, T, Filho, BP, Cortez, DA and Nakamura, CV. (2012) Intramuscular and topical treatment of cutaneous leishmaniasis lesions in mice infected with Leishmania amazonensis using coumarin (-) mammea A/BB. Phytomedicine 19(13), 11961199. https://doi.org/10.1016/j.phymed.2012.08.001Google Scholar
Rodríguez-Hernández, KD, Martínez, I, Reyes-Chilpa, R and Espinoza, B (2020) Mammea type coumarins isolated from Calophyllum brasiliense induced apoptotic cell death of Trypanosoma cruzi through mitochondrial dysfunction, ROS production and cell cycle alterations. Bioorganic Chemistry 100, 103894. https://doi.org/10.1016/j.bioorg.2020.103894Google Scholar
Ferreira, ME, de Arias, AR, Yaluff, G, de Bilbao, NV, Nakayama, H, Torres, S,…Fournet, A (2010) Anti-leishmanial activity of furoquinolines and coumarins from Helietta apiculata. Phytomedicine 17(5), 375378. https://doi.org/10.1016/j.phymed.2009.09.009Google Scholar
Kermani, EK, Sajjadi, SE, Hejazi, SH, Arjmand, R, Saberi, S and Eskandarian, AA (2016) Anti-Leishmania activity of osthole. Pharmacognosy Research 8 (Suppl 1), S14. https://doi.org/10.4103/0974-8490.178650Google Scholar
Napolitano, HB, Silva, M, Ellena, J, Rodrigues, BD, Almeida, AL, Vieira, PC,…Thiemann, OH (2004) Aurapten, a coumarin with growth inhibition against Leishmania major promastigotes. Brazilian Journal of Medical and Biological Research 37(12), 18471852. https://doi.org/10.1590/s0100-879x2004001200010Google Scholar
Gonçalves, GA, Spillere, AR, das Neves, GM, Kagami, LP, von Poser, GL, Canto, RFS and Eifler-Lima, V (2020) Natural and synthetic coumarins as anti-leishmanial agents: A review. European Journal of Medicinal Chemistry 203, 112514. https://doi.org/10.1016/j.ejmech.2020.112514Google Scholar
Bisacchi, GS (2015) Origins of the quinolone class of antibacterials: An expanded "Discovery Story”. Journal of Medicinal Chemistry 58(12), 48744882. https://doi.org/10.1021/jm501881cGoogle Scholar
Yun, D, Yoon, SY, Park, SJ and Park, YJ (2021) The anticancer effect of natural plant alkaloid isoquinolines. International Journal of Molecular Sciences 22(4). https://doi.org/10.3390/ijms22041653Google Scholar
Shang, XF, Morris-Natschke, SL, Liu, YQ, Guo, X, Xu, XS, Goto, M,…Lee, KH (2018) Biologically active quinoline and quinazoline alkaloids part I. Medicinal Research Reviews 38(3), 775828. https://doi.org/10.1002/med.21466Google Scholar
Heeb, S, Fletcher, MP, Chhabra, SR, Diggle, SP, Williams, P and Cámara, M (2011) Quinolones: From antibiotics to autoinducers. FEMS Microbiology Reviews 35(2), 247274. https://doi.org/10.1111/j.1574-6976.2010.00247.xGoogle Scholar
Pham, TDM, Ziora, ZM and Blaskovich, MAT (2019) Quinolone antibiotics. Medchemcomm 10(10), 17191739. https://doi.org/10.1039/c9md00120dGoogle Scholar
McConnell, EV, Bruzual, I, Pou, S, Winter, R, Dodean, RA, Smilkstein, MJ,…Doggett, JS (2018) Targeted structure-activity analysis of endochin-like quinolones reveals potent qi and qo site inhibitors of Toxoplasma gondii and Plasmodium falciparum cytochrome bc. ACS Infectious Diseases Journal 4(11), 15741584. https://doi.org/10.1021/acsinfecdis.8b00133Google Scholar
Ortiz, D, Forquer, I, Boitz, J, Soysa, R, Elya, C, Fulwiler, A,…Landfear, SM (2016). Targeting the cytochrome bc1 complex of Leishmania parasites for discovery of novel Drugs. Antimicrobial Agents and Chemotherapy 60(8), 49724982. https://doi.org/10.1128/AAC.00850-16Google Scholar
Coimbra, ES, Antinarelli, LM, Silva, NP, Souza, IO, Meinel, RS, Rocha, MN,…da Silva, AD (2016) Quinoline derivatives: Synthesis, leishmanicidal activity and involvement of mitochondrial oxidative stress as mechanism of action. Chemico-Biological Interactions 260, 5057. https://doi.org/10.1016/j.cbi.2016.10.017Google Scholar
Calla-Magariños, J, Quispe, T, Giménez, A, Freysdottir, J, Troye-Blomberg, M and Fernández, C (2013) Quinolinic alkaloids from Galipea longiflora Krause suppress production of proinflammatory cytokines in vitro and control inflammation in vivo upon Leishmania infection in mice. Scandinavian Journal of Immunology 77(1), 3038. https://doi.org/10.1111/sji.12010Google Scholar
Torres Suarez, E, Granados-Falla, DS, Robledo, SM, Murillo, J, Upegui, Y and Delgado, G (2020) Anti-leishmanial activity of synthetic analogs of the naturally occurring quinolone alkaloid N-methyl-8-methoxyflindersin. PLoS One 15(12), e0243392. https://doi.org/10.1371/journal.pone.0243392Google Scholar
Coy Barrera, CA, Coy Barrera, ED, Granados Falla, DS, Delgado Murcia, G and Cuca Suarez, LE (2011) Seco-limonoids and quinoline alkaloids from Raputia heptaphylla and their anti-leishmanial activity. Chemical and Pharmaceutical Bulletin 59(7), 855859. https://doi.org/10.1248/cpb.59.855Google Scholar
Croft, SL, Hogg, J, Gutteridge, WE, Hudson, AT and Randall, AW (1992) The activity of hydroxynaphthoquinones against Leishmania donovani. Journal of Antimicrobial Chemotherapy 30(6), 827832. https://doi.org/10.1093/jac/30.6.827Google Scholar
Luque-Ortega, JR, Rivero-Lezcano, OM, Croft, SL and Rivas, L (2001 ) In vivo monitoring of intracellular ATP levels in Leishmania donovani promastigotes as a rapid method to screen drugs targeting bioenergetic metabolism. Antimicrobial Agents and Chemotherapy 45(4), 11211125. https://doi.org/10.1128/AAC.45.4.1121-1125.2001Google Scholar
Smith, L, Serrano, DR, Mauger, M, Bolás-Fernández, F, Dea-Ayuela, MA and Lalatsa, A (2018) Orally bioavailable and effective buparvaquone lipid-based nanomedicines for visceral leishmaniasis. Molecular Pharmaceutics 15(7), 25702583. https://doi.org/10.1021/acs.molpharmaceut.8b00097Google Scholar
Monteiro, LM, Löbenberg, R, Barbosa, EJ, de Araujo, GLB, Sato, PK, Kanashiro, E,…Bou Chacra, NA (2022) Oral administration of buparvaquone nanostructured lipid carrier enables in vivo activity against Leishmania infantum. European Journal of Pharmaceutical Sciences 169, 106097. https://doi.org/10.1016/j.ejps.2021.106097Google Scholar
Thapa, R, Mondal, S, Riikonen, J, Rantanen, J, Näkki, S, Nissinen, T,…Lehto, VP (2021) Biogenic nanoporous silicon carrier improves the efficacy of buparvaquone against resistant visceral leishmaniasis. PLOS Neglected Tropical Diseases 15(6), e0009533. https://doi.org/10.1371/journal.pntd.0009533Google Scholar
Qi, W, Xiong, D and Long, M (2022) Quercetin: Its Antioxidant Mechanism, Antibacterial Properties and Potential Application in Prevention and Control of Toxipathy. Molecules 27(19), 6545. https://doi.org/10.3390/molecules27196545Google Scholar
Ghareeb, MA, Zayan, AZ, Shari, FH, and Sayed, AM (2024) Unveiling the potential quercetin: chemistry, health benefits. In Osredkar, J (ed.), Toxicity, and Cutting-Edge Advances in Quercetin – Effects on Human Health. Publisher, IntechOpen. https://doi.org/10.5772/intechopen.1005344Google Scholar
Deepika, and Maurya, PK (2022) Health benefits of quercetin in Age-related diseases. Molecules 27(8) 2498. https://doi.org/10.3390/molecules27082498Google Scholar
Sarkar, S, Mandal, S, Sinha, J, Mukhopadhyay, S, Das, N and Basu, MK (2002) Quercetin: evaluation as an anti-leishmanial agent in vivo in hamsters using different vesicular delivery modes. Journal of Drug Targeting 10(8), 573578. https://doi.org/10.1080/106118021000072681Google Scholar
Sousa-Batista, AJ, Poletto, FS, Philipon, CIMS, Guterres, SS, Pohlmann, AR, Bergmann, B (2017) Lipid-core nanocapsules increase the oral efficacy of quercetin in cutaneous leishmaniasis. Parasitology 144(13), 17691774. https://doi.org/10.1017/S003118201700097XGoogle Scholar
Mehwish, S, Varikuti, S, Ali Khan, M, Khan, T, Khan, IU, Satoskar, A,…Ullah, N (2021) Bioflavonoid-Induced Apoptosis and DNA damage in amastigotes and promastigotes of Leishmania donovani: Deciphering the mode of action. Molecules 26(19), 5843. https://doi.org/10.3390/molecules26195843Google Scholar
Cataneo, AHD, Tomiotto-Pellissier, F, Miranda-Sapla, MM, Assolini, JP, Panis, C, Kian, D,…Pavanelli, WR (2019) Quercetin promotes antipromastigote effect by increasing the ROS production and anti-amastigote by upregulating Nrf2/HO-1 expression, affecting iron availability. Biomedicine & Pharmacotherapy 113, 108745. https://doi.org/10.1016/j.biopha.2019.108745Google Scholar
Fonseca-Silva, F, Inacio, JD, Canto-Cavalheiro, MM and Almeida-Amaral, EE (2011) Reactive oxygen species production and mitochondrial dysfunction contribute to quercetin induced death in Leishmania amazonensis. PLoS One 6(2), e14666. https://doi.org/10.1371/journal.pone.0014666Google Scholar
Muzitano, MF, Falcão, CA, Cruz, EA, Bergonzi, MC, Bilia, AR, Vincieri, FF,…Costa, SS. (2009) Oral metabolism and efficacy of Kalanchoe pinnata flavonoids in a murine model of cutaneous leishmaniasis. Planta Medica 75(4), 307311. https://doi.org/10.1055/s-0028-1088382Google Scholar
Dos Santos, RF, Da Silva, T, Brito, ACS, Inácio, JD, Ventura, BD, Mendes, MAP,…Da Silva, SAG (2022) Therapeutic effect of oral quercetin in hamsters infected with. Frontiers in Cellular and Infection Microbiology 12, 1059168. https://doi.org/10.3389/fcimb.2022.1059168Google Scholar
Ribeiro-Romão, RP, Moreira, OC, Osorio, EY, Cysne-Finkelstein, L, Gomes-Silva, A, Valverde, JG,…Pinto, EF (2014) Comparative evaluation of lesion development, tissue damage, and cytokine expression in golden hamsters (Mesocricetus auratus) infected by inocula with different Leishmania (Viannia) braziliensis concentrations. Infection and Immunity 82(12), 52035213. https://doi.org/10.1128/IAI.02083-14Google Scholar
Carrillo-Garmendia, A, Madrigal-Perez, LA and Regalado-Gonzalez, C (2023) The multifaceted role of quercetin derived from its mitochondrial mechanism. Molecular and Cellular Biochemistry. https://doi.org/10.1007/s11010-023-04833-wGoogle Scholar
da Silva, JM, Antinarelli, LM, Ribeiro, A, Coimbra, ES and Scio, E (2015) The effect of the phytol-rich fraction from Lacistema pubescens against Leishmania amazonensis is mediated by mitochondrial dysfunction. Experimental Parasitology 159, 143150. https://doi.org/10.1016/j.exppara.2015.09.009Google Scholar
Monzote, L, Lackova, A, Staniek, K, Steinbauer, S, Pichler, G, Jäger, W and Gille, L (2017) The anti-leishmanial activity of xanthohumol is mediated by mitochondrial inhibition. Parasitology 144(6), 747759. https://doi.org/10.1017/S0031182016002389Google Scholar
Dighal, A ;De Sarkar, S ;Gille, L ;Chatterjee, M (2021) Can the iron content of culture media impact on the leishmanicidal effect of artemisinin? Free Radical Research 55 (3) 282295. https://doi.org/10.1080/10715762.2021.1939325Google Scholar
Lazarin-Bidóia, D, Garcia, FP, Ueda-Nakamura, T, Silva, SO and Nakamura, CV (2022) Natural compounds based chemotherapeutic against Chagas disease and Leishmaniasis: Mitochondrion as a strategic target. Memórias do Instituto Oswaldo Cruz 117, e220396. https://doi.org/10.1590/0074-02760220396Google Scholar
De Sarkar, S, Sarkar, D, Sarkar, A, Dighal, A, Staniek, K, Gille, L and Chatterjee, M (2019) Berberine chloride mediates its anti-leishmanial activity by inhibiting Leishmania mitochondria. Parasitology Research 118(1), 335345. https://doi.org/10.1007/s00436-018-6157-3Google Scholar
Awasthi, BP, Kathuria, M, Pant, G, Kumari, N and Mitra, K (2016) Plumbagin, a plant-derived naphthoquinone metabolite induces mitochondria mediated apoptosis-like cell death in Leishmania donovani: an ultrastructural and physiological study. Apoptosis 21(8), 941953. https://doi.org/10.1007/s10495-016-1259-9Google Scholar
Upegui Zapata, YA, Echeverri, F, Quiñones, W, Torres, F, Nacher, M, Rivas, LI,…Robledo, SM (2020) Mode of action of a formulation containing hydrazones and saponins against Leishmania spp. Role in mitochondria, proteases and reinfection process. International Journal for Parasitology: Drugs and Drug Resistance 13, 94106. https://doi.org/10.1016/j.ijpddr.2020.06.004Google Scholar
Pereira, KLG, Vasconcelos, NBR, Braz, JVC, InÁcio, JDF, Estevam, CS, Correa, CB,…Scher, R (2020) Ethanolic extract of Croton blanchetianus ball induces mitochondrial defects in Leishmania amazonensis promastigotes. Anais da Academia Brasileira de Ciências 92 (suppl 2), e20180968. https://doi.org/10.1590/0001-3765202020180968Google Scholar
Dagnino, APA, Mesquita, CS, Dorneles, GP, Teixeira, VON, de Barros, FMC, Vidal Cana-Capatinta, G,…Romão, PRT (2018) Phloroglucinol derivatives from Hypericum species trigger mitochondrial dysfunction in Leishmania amazonensis. Parasitology 145(9), 11991209. https://doi.org/10.1017/S0031182018000203Google Scholar
Fonseca-Silva, F, Canto-Cavalheiro, MM, Menna-Barreto, RF and Almeida-Amaral, EE (2015). Effect of apigenin on Leishmania amazonensis is associated with reactive oxygen species production followed by mitochondrial dysfunction. Journal of Natural Products 78(4), 880884. https://doi.org/10.1021/acs.jnatprod.5b00011Google Scholar
Elmahallawy, EK, Jiménez-Aranda, A, Martínez, AS, Rodriguez-Granger, J, Navarro-Alarcón, M, Gutiérrez-Fernández, J and Agil, A (2014) Activity of melatonin against Leishmania infantum promastigotes by mitochondrial dependent pathway. Chemico-Biological Interactions 220, 8493. https://doi.org/10.1016/j.cbi.2014.06.016Google Scholar
Bortoleti, BTDS, Tomiotto-Pellissier, F, Gonçalves, MD, Miranda-Sapla, MM, Assolini, JP, Carloto, AC,…Pavanelli, WR (2019) Caffeic acid has antipromastigote activity by apoptosis-like process; and anti-amastigote by TNF-α/ROS/NO production and decreased of iron availability. Phytomedicine 57, 262270. https://doi.org/10.1016/j.phymed.2018.12.035Google Scholar
Das, R, Roy, A, Dutta, N and Majumder, HK (2008) Reactive oxygen species and imbalance of calcium homeostasis contributes to curcumin induced programmed cell death in Leishmania donovani. Apoptosis 13(7), 867882. https://doi.org/10.1007/s10495-008-0224-7Google Scholar
Jung, KH., Lee, JH., Park, JW, Moon, SH, Cho, YS, Choe, YS and Lee, KH (2016) Effects of curcumin on cancer cell mitochondrial function and potential monitoring with 18F-FDG uptake. Oncology Reports 35(2), 861868. https://doi.org/10.3892/or.2015.4460Google Scholar
Chaughule, RS and Barve, RS (2023) Role of herbal medicines in the treatment of infectious diseases. Vegetos 111. https://doi.org/10.1007/s42535-022-00549-2Google Scholar
Dong, M, Zheng, G, Gao, F, Li, M and Zhong, C (2022) Three-carbon linked dihydroartemisinin-isatin hybrids: Design, synthesis and their antiproliferative anticancer activity. Frontiers in Pharmacology 13, 834317. https://doi.org/10.3389/fphar.2022.834317Google Scholar
Guan, L, Wang, H, Xu, X and Fan, H (2023) Therapeutical utilization and repurposing of artemisinin and its derivatives: A narrative review. Advanced Biology (Weinh), 7(8), e2300086. https://doi.org/10.1002/adbi.202300086Google Scholar
Dong, Y, Liu, L, Han, J, Zhang, L, Wang, Y, Li, J,…Hu, X (2022) Worldwide research trends on artemisinin: A bibliometric analysis from 2000 to 2021. Frontiers in Medicine (Lausanne) 9, 868087. https://doi.org/10.3389/fmed.2022.868087Google Scholar
Kamat, S and Kumari, M (2021) Repurposing chloroquine against multiple diseases with special attention to SARS-CoV-2 and associated toxicity. Frontiers in Pharmacology 12, 576093. https://doi.org/10.3389/fphar.2021.576093Google Scholar
Jeena, MT, Kim, S, Jin, S and Ryu, JH (2019) Recent progress in mitochondria-targeted drug and drug-free agents for cancer therapy. Cancers (Basel) 12(1), 4. https://doi.org/10.3390/cancers12010004Google Scholar
Xu, C, Xiao, L, Zhang, X, Zhuang, T, Mu, L and Yang, X (2021) Synthesis and biological activities of novel mitochondria-targeted artemisinin ester derivatives. Bioorganic & Medicinal Chemistry Letters 39, 127912. https://doi.org/10.1016/j.bmcl.2021.127912Google Scholar
Scotti, L, Ishiki, H, Mendonça Júnior, FJ, Da Silva, MS and Scotti, MT (2015) In-silico analyses of natural products on Leishmania enzyme targets. Mini Reviews in Medicinal Chemistry, 15(3), 253269. https://doi.org/10.2174/138955751503150312141854Google Scholar
Bora, N and Jha, AN (2020) Metabolic pathway analysis identifying target against leishmaniasis – A kinetic modeling approach. Frontiers in Genetics 11, 179. https://doi.org/10.3389/fgene.2020.00179Google Scholar
Chávez-Fumagalli, MA, Lage, DP, Tavares, GSV, Mendonça, DVC, Dias, DS, Ribeiro, PAF,…Coelho, EAF (2019) In silico Leishmania proteome mining applied to identify drug target potential to be used to treat against visceral and tegumentary leishmaniasis. Journal of Molecular Graphics and Modelling 87, 8997. https://doi.org/10.1016/j.jmgm.2018.11.014Google Scholar
Kant, V, Kumar, P, Ranjan, R, Mandal, D and Vijayakumar, S (2022) In silico screening, molecular dynamic simulations, and in vitro activity of selected natural compounds as an inhibitor of Leishmania donovani 3-mercaptopyruvate sulfurtransferase. Parasitology Research 121(7), 20932109. https://doi.org/10.1007/s00436-022-07532-5Google Scholar
Khandibharad, S and Singh, S (2024) Synthetic biology for combating leishmaniasis. Frontiers in Microbiology 15, 1338749. https://doi.org/10.3389/fmicb.2024.1338749Google Scholar
Challapa-Mamani, MR, Tomás-Alvarado, E, Espinoza-Baigorria, A, León-Figueroa, DA, Sah, R, Rodriguez-Morales, AJ and Barboza, JJ (2023) Molecular docking and molecular dynamics simulations in related to Leishmania donovani: An update and literature review. Tropical Medicine and Infectious Disease 8(10), 457. https://doi.org/10.3390/tropicalmed8100457Google Scholar
Figure 0

Figure 1. Distribution of leishmaniasis in the Old World and the New World. The figure shows species distribution of the Leishmania parasite in the old world and the new world (Ref 16). CL: cutaneous leishmaniasis; MCL: mucocutaneous leishmaniasis; VL: visceral leishmaniasis.

Figure 1

Figure 2. Life cycle of the Leishmania parasite. (1) Sandfly, the invertebrate host, releases infective metacyclic promastigotes in the mammalian bloodstream while taking a blood meal. The parasites enter an environment of 37°C in the mammalian skin from where they are picked up by the phagocytic cells. (2) The promastigotes convert to amastigotes and proliferate within the phagolysosomes of the mammalian macrophages. (3) Amastigotes are released from the macrophages when saturating numbers are reached within a cell. (4) Amastigotes and parasitized macrophages are picked up by the sandfly during a bite. (5) Amastigotes convert to promastigotes within the gut of the sandfly and the infective metacyclic forms move to the proboscis to be delivered to the mammalian bloodstream during a bite (Ref 14).

Figure 2

Figure 3. The electron transport chain. Complex I (NADH ubiquinone oxidoreductase), complex II (succinate ubiquinone oxidoreductase); complex III (cytochrome c3+ oxidoreductase); complex IV (cytochrome C oxidase with ubiquinone) are located on the inner mitochondrial membrane as integral membrane proteins. Complex I transfers two electrons to ubiquinone when a simultaneous translocation of protons occurs. Coenzyme Q and cytochrome C serve as mobile electron carriers to facilitate the production of ATP through oxidative phosphorylation. CoQ10 allows the transfer of electrons to complex III which transfers these electrons to the cytochrome C responsible for connecting to complex IV where the reduction of O2 to H2O will take place. Complexes I, III, and IV function as proton pumps. The generation of an electrochemical gradient between the intermembrane space and the mitochondrial matrix occurs because of the pumping of protons. The ΔΨm thus produced drives ATP synthesis from ADP and inorganic phosphate by the F1F0 ATP synthase (Refs 35, 36). Cyt C: cytochrome c; IMM: inner mitochondrial membrane; IMS: Intermembrane space; OMM: Outer mitochondrial membrane; NADH: nicotinamide adenine dinucleotide + hydrogen.

Figure 3

Figure 4. In vivo tested plant-based possible anti-leishmanials that could target mitochondria. Artemisinin, chloroquine, quercetin, coumarin and R1–R5 quinolone are shown.

Figure 4

Table 1 Table describing the plants from which the compounds were first prepared, the part of the plants from where the extracts were made, and the targets of the compounds on the parasite mitochondrion