Next Article in Journal
Detection of Aromatic Hydrocarbons in Aqueous Solutions Using Quartz Tuning Fork Sensors Modified with Calix[4]arene Methoxy Ester Self-Assembled Monolayers: Experimental and Density Functional Theory Study
Previous Article in Journal
Ultra-High Cycling Stability of 3D Flower-like Ce(COOH)3 for Supercapacitor Electrode via a Facile and Scalable Strategy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Natural Products in the Treatment of Schistosomiasis

by
Carine Machado Azevedo
1,
Cássio Santana Meira
1,2,
Jaqueline Wang da Silva
2,
Danielle Maria Nascimento Moura
3,
Sheilla Andrade de Oliveira
3,
Cícero Jádson da Costa
3,
Emanuelle de Souza Santos
2 and
Milena Botelho Pereira Soares
1,2,*
1
Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Brazil
2
SENAI Institute of Innovation in Health Advanced Systems (CIMATEC ISI SAS), University Center SENAI/CIMATEC, Salvador 41650-010, Brazil
3
Aggeu Magalhães Institute, Oswaldo Cruz Foundation (IAM-FIOCRUZ/PE), Recife 50740-465, Brazil
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(19), 6807; https://doi.org/10.3390/molecules28196807
Submission received: 29 April 2023 / Revised: 2 August 2023 / Accepted: 3 August 2023 / Published: 26 September 2023
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
It is estimated that 250 million people worldwide are affected by schistosomiasis. Disease transmission is related to the poor sanitation and hygiene habits that affect residents of impoverished regions in tropical and subtropical countries. The main species responsible for causing disease in humans are Schistosoma Mansoni, S. japonicum, and S. haematobium, each with different geographic distributions. Praziquantel is the drug predominantly used to treat this disease, which offers low effectiveness against immature and juvenile parasite forms. In addition, reports of drug resistance prompt the development of novel therapeutic approaches. Natural products represent an important source of new compounds, especially those obtained from plant sources. This review compiles data from several in vitro and in vivo studies evaluating various compounds and essential oils derived from plants with cercaricidal and molluscicidal activities against both juvenile and adult forms of the parasite. Finally, this review provides an important discussion on recent advances in molecular and computational tools deemed fundamental for more rapid and effective screening of new compounds, allowing for the optimization of time and resources.

1. Introduction

Schistosomiasis, a neglected tropical disease (NTD), infects around 250 million people worldwide [1,2]. Like other NTDs, it predominantly affects the poorer populations living in tropical and subtropical regions [2,3]. There are three main species of trematodes of the genus Schistosoma, the causative agents of the disease in humans: S. mansoni and S. japonicum (which cause intestinal and hepatosplenic disease) and S. haematobium (which affects the genitourinary system) [4]. The distribution of each species follows that of its respective intermediate host snail. In Africa and the Middle East, S. haematobium and S. mansoni are found, while the latter is also present in South America and the West Indies. However, S. japonicum is endemic to some Asian countries, such as China, the Philippines, Indonesia, and the Mekong Delta region [3]. Although other species, such as S. intercalatum, S. guineensis, and S. mekongi, are also known to cause the disease in humans, their importance is secondary since distribution is restricted to Central and West Africa (the first two species) and Laos and eastern Cambodia [2].
Infection occurs when an individual comes into contact with water containing cercariae released by intermediate host snails, which may be of the genus Biomphalaria (for S. mansoni), Oncomelania (for S. japonicum), or Bulinus (for S. haematobium). While cercariae can survive in water for 1 to 3 days [5], the ability to infect reduces rapidly within a few hours of release [6]. After penetrating the human host’s skin, the parasites circulate, reaching the lungs and then the liver where they transform into young worms or schistosomula. Within 4 to 6 weeks, the worms mature within the portal venous system, mate, and migrate in pairs to the mesenteric veins [5], in the case of S. mansoni and S. japonicum, or to the vesical venous plexus of the urogenital system, in the case of S. haematobium [3].
After mating, females begin to lay eggs that may be retained in the liver (forming granulomas and subsequent fibrosis) or pass through the intestine, returning to the environment along with feces. In the case of S. haematobium, eggs are excreted through the urine. The lifespan of adult worms generally ranges from 3 to 5 years, but reports have indicated survival for up to 30 years [5]. Eggs, upon coming into contact with water, hatch and release the miracidia that infect snails, within which asexual replication leads to the formation of the mother and offspring sporocysts, in addition to subsequent cercariae [5,7]. Around 28 to 30 days (S. mansoni and S. haematobium) or 90 days (S. japonicum) following infection, the snail begins to shed cercariae [3]. This process is stimulated by light, mainly occurring during the day [5].
Since infection occurs through contact with water contaminated by cercariae, poor sanitation and hygiene can place children, adolescents, and adults at risk [8]. Contamination occurs mainly in poor regions of developing countries, in which the prevalence of disease is higher [3]. Other activities involving contact with water, i.e., domestic (washing clothes and dishes in open bodies of freshwater), recreational (bathing in rivers and lakes), or professional activities can expose people to risk [8]. In this context, children and pregnant women are those most susceptible to infection and reinfection. In children, schistosomiasis can cause malnutrition, reduced growth, and cognitive impairment, while in pregnant women it can lead to premature birth, low birth weight, and higher maternal morbidity and mortality [9].
The pathology of schistosomiasis is closely related to the large quantity of eggs deposited by females, which accumulate in the liver/intestine or bladder/urogenital system [2,10]. However, studies have shown that host genetic factors may also be associated with more severe manifestations of disease (such as HLA class I and class II antigens) or with protection against severe hepatic fibrosis (such as HLA-DP alleles). In addition, the development of resistance to reinfection with S. mansoni has been linked to the SM1 locus located on chromosome 5q31–q33 [11]. The disease is characterized by three distinct phases: acute (following primary infection, more common in travelers or immigrants to endemic areas), established, and late chronic (commonly observed in individuals living in endemic areas) [2]. The acute phase (also known as Katayama fever or Katayama syndrome) commonly occurs in persons infected for the first time, with the symptoms exhibited in response to antigens released by schistosomula during the migration process, as well as by recently laid eggs [2,8]. This phase occurs between 2 weeks to 3 months after exposure to cercariae, with a typical clinical presentation consisting of fever, myalgia, headache, bloody diarrhea, hepatosplenomegaly, eosinophilia, non-productive cough, patchy infiltrates on chest radiography, and elevated IgE levels [2,3,5,10]. Residents in endemic areas do not exhibit an acute symptomatic phase, and usually develop an established active infection characterized by the presence of adult worms that produce eggs that are excreted in feces or urine. Although the adult worms present in blood vessels do not provoke an inflammatory reaction, eggs produce soluble antigens that induce the formation of granulomas that lead to tissue fibrosis. In the chronic phase, the accumulation of numerous granulomas provokes the formation of periportal fibrosis in the liver (S. mansoni and S. japonicum), leading to portal hypertension and the appearance of esophageal and gastric varices. These varicose veins can rupture and cause bleeding, potentially resulting in consequent death [3]. In the case of S. haematobium, patients present obstructive disease in the urinary and reproductive system, in addition to bladder calcification, genital lesions, kidney involvement, i.e., hydronephrosis and renal failure, and bladder cancer [3].
As neglected diseases, such as schistosomiasis, are not prioritized by pharmaceutical companies, therapeutic options are limited, outdated, and sometimes even non-existent. Despite being a disease that has been studied for several decades, the only effective treatment option against different species of Schistosoma is praziquantel (PZQ), which was discovered in 1972 and has been available for treatment since the 1980s [12]. In accordance with WHO recommendations, preventive chemotherapy involving PZQ is indicated for affected populations and at-risk groups. The frequency of treatment is determined by the prevalence of infection in school-aged children [13]. While the anthelmintic activity of PZQ remains uncertain, some authors speculate that it likely inhibits the Na+ and K+ pump in adult worms, increasing the permeability of the helminth membrane to certain monovalent and bivalent cations, such as calcium, which then leads to the intensification of muscle activity, followed by contraction and spastic paralysis [14,15]. While this drug achieves a cure rate of between 60 and 90%, it lacks activity against immature and juvenile parasites, which have been shown to survive drug exposure [3]. Consequently, new adult worms may appear 1 to 2 months after treatment, since administration consists of a single dose [2]. Other disadvantages involving the use of PZQ are related to its racemic nature, as only half of the dose is pharmacologically utilized and what is absorbed becomes rapidly metabolized into inactive metabolites, resulting in relatively minimal drug contact with parasites in the host’s bloodstream [16,17]. Despite advantages of low cost and facile administration in adults (single oral dose of 40 mg/kg) [9], there is no pediatric formulation for PZQ, the pills are large, and a bitter taste may further contribute to the low cure rates observed among preschool children [9,10]. Another drug, oxamniquine, which has demonstrated efficacy only against S. mansoni, is restricted to use in South America. Like praziquantel, treatment is administered orally via a single dose and has few reported side effects. In sum, the fact that commercially available drugs do not prevent reinfection, coupled with reports of drug resistance, making the search for alternative chemotherapeutic solutions to overcome current limitations urgent, whether through the development of new drugs or combination therapy involving PZQ [9].
In this context, natural products, which have been used in the treatment of human diseases for many years, are produced by living organisms, such as plants, animals, microbes, and marine organisms [18]. When derived from plants, natural substances are usually obtained from leaves, bark, stems, or roots, and may include alkaloids, phenolic compounds, flavonoids, terpenoids, tannins, saponins, and steroids [19]. Several diseases (such as malaria, dyspepsia, liver disorders, and glaucoma) are currently treated with drugs produced from plant-based bioactive entities, e.g., quinine, silymarin, artemisinin, or pilocarpine, among others [20]. Moreover, around 60% of the antiparasitic compounds used between 1981 and 2014 originated from natural products [18]. This study presents an overview of the anthelmintic activity exhibited by several natural products, including essential oils, in different in vitro and in vivo systems. In addition, we also describe useful tools for discovering new drugs that can enable reductions in both time and cost.

2. Plant-Derived Compounds

Despite the pharmaceutical industry’s growing interest in synthetic molecules for drug development, natural products remain a valuable source for new molecule discovery, including those with antiparasitic activity [21]. In this context, several reports have explored well-known natural products (e.g., quercetin, curcumin, pirplatine, and others), novel natural molecules [22,23,24] (Figure 1) and essential oils and their components [25] in the search for new antischistosomal drugs.

2.1. In Vitro Studies

Several in vitro studies (Table 1) have demonstrated the antischistosomal activity of a variety of natural molecules, especially against adult worms of S. mansoni. These reports mainly describe the ability of natural products to decrease worm motility and induce death via different pathways (Figure 2). The terpene nerolidol (3,7,11-trimethyl-1,6,10-dodecatrien-3-ol), also known as peruviol, was shown to promote a reduction in worm motility as well as death in adult parasites at concentrations ranging between 62.5 and 250 µM after 48 or 72 h of treatment; moreover, adult male parasites were found to be more susceptible to nerolidol than female worms [26]. In another investigation involving 10 triterpenes with the cucurbitane skeleton, balsaminol F and karavilagenin were identified as promising antischistosomal agents, since motor activity became significantly reduced (at 10–50 µM) and 100% death was observed in adult worms of S. mansoni at 100 µM with LC50 values of 14.7 and 28.9 µM respectively against 56-day-old adult S. mansoni [27]. In addition, licochalcone A, a characteristic chalcone of licorice, presented LC50 values of 9.12 and 9.52 µM against female and male adult worms, respectively, with impairment of motor activity observed at concentrations between 12.5 and 200 µM [28]. In fact, most of the compounds described in Table 1, especially those belonging to the terpene and chalcone classes, have been shown to reduce motility and induce death in adult S. mansoni worms [2,22,29,30,31,32,33,34,35,36,37,38,39].
Interestingly, the changes observed in motility and death of adult S. mansoni worms caused by the natural products described in Table 1 are mainly associated with alterations in the tegument [26,29,30,34,36,43,44,45,46,48]. The tegument is considered a key structure in the evasion of host immune response, acquiring nutrients, excreting catabolic products, and targeting drug absorption, among other physiological processes [49]. Therefore, it is an attractive target for the development of antischistosomal drugs. A natural product featuring the tegument of S. mansoni as an already characterized target is diterpene phytol. This natural product, at concentrations between 50 to 150 µg/mL, promotes severe tegument damage in schistosomes, such as body deformation, morphological disfiguring of the oral and ventral suckers, extensive sloughing, loss of tubercles, and shrinking [35,44]. Moreover, quantitative analysis revealed a concentration-dependent reduction in the number of intact tubercles after phytol treatment, with complete tubercle destruction observed at 100 µg/mL [44]. A reduction in the number of intact tubercles and morphological tegument alterations was also observed in adult S. mansoni worms treated with chalcones, especially licochalcone A and licoflavone B [33,36,48]. Transmission electron microscopy was used to visualize the formation of vacuoles of different sizes in the tegument and swelling in different regions of the integument due to the presence of sparse matrix after treatment with 10 µM of licochalcone A. In addition, licochalcone A also promoted swelling and degeneration of the mitochondria, as well as nuclear chromatin condensation, which were all correlated with increased superoxide anion levels and decreased superoxide dismutase activity. Interestingly, licochalcone A presented schistosomicidal activity without affecting the viability of mammalian cells (CHO-K1 cells; Chinese hamster ovary fibroblasts) at concentrations ≤400 µM [48].
Similarly, licoflavone B treatment caused massive disintegration of the tegumental surface in association with disruption of tubercles. These effects were accompanied by a pronounced inhibition of S. mansoni ATPase and ADPase activity, with resulting IC50 values of 23.78 and 31.51 µM, respectively, which was corroborated by docking studies involving licoflavone B and SmATPDase 1 [33]. The schistosomicidal activity of licoflavone B was observed at concentrations (25–200 µM) nontoxic to mammalian Vero cells [33]. Similar results were described by Pereira et al. [36], who used S. mansoni ATP diphosphohydrolases as a target and identified schistosomicidal activity in a series of chalcones without affecting cell viability [36].
Some natural products were also shown to interfere with the reproductive fitness of S. mansoni. Regarding oviposition, licoflavone B reduced the total number of eggs laid at sub-lethal concentrations (2.5, 5, and 10 µM) and inhibited 100% of egg-laying at 10 µM [33]. Oviposition was also affected by other natural products, such as dermaseptin 01, balsaminol F, karavilagenin C, phytol, dibenzylbutyrolactonic lignans, and curcumin [24,27,29,37,44].
Finally, some natural products demonstrated activity against S. mansoni cercariae (infective larval stage) as well as snails (Table 1) [24,32,47]. The alkaloid diethyl 4-phenyl-2,6-dimethyl-3,5-pyridinedicarboxylate exhibited potent cercaricidal activity (LC100 = 2 μg/mL) in addition to activity against adult B. glabrata (LC90 = 36.43 μg/mL) [33]. Barbatic acid, a lichen metabolite, provoked substantial molluscicidal activity against snails at concentrations ranging between 10.5 and 50 µg/mL, with optimal effects (100% lethality) observed at 25 µg/mL. In addition, barbatic acid also presented cercaricidal activity, completely eliminating cercariae at concentrations between 1 and 100 µg/mL after 60 min of drug exposure. Importantly, both cercaricidal and molluscicidal activity was found to occur at concentrations that did not alter Artemia salina viability [47]. Lastly, it is important to highlight the in vitro activity of curcumin. At different treatment times, LC50 values below 10 ug/mL demonstrated efficacy against cercariae and inhibited ability egg-laying capacity as well as egg hatchability, causing death in newborns, embryos, and adult B. glabrata snails [24].

2.2. In Vivo Studies

The antischistosomal effect of orally or intraperitoneally administered natural compounds has also been observed in several in vivo studies involving mice (Table 2). Figure 3 details the main outcomes identified in this literature review.
Initial work by Allam (2009) [50] demonstrated the anti-schistosomal activity of curcumin (400 mg/kg) in a murine model through the modulation of both cellular and humoral immune response. The intraperitoneal treatment of infected mice with curcumin resulted in a significant reduction in levels of interleukin (IL)-12 and tumor necrosis factor alpha (TNF-a) compared to the untreated infected group. This downregulation can be attributed to curcumin’s inhibition of nuclear factor kappa B (NF-κB), leading to a subsequent decrease in pro-inflammatory cytokines [68]. Moreover, the treated animals exhibited elevated levels of specific IgG and IgG1 antibodies against soluble worm and soluble egg antigens. The hepatoprotective property of curcumin has been noted in the context of liver fibrosis [51]. El-Agamy et al., 2011, [51] also reported similar antifibrotic effects in S. mansoni infection following oral curcumin treatment (300 mg/kg/day) for 2 weeks.
Interestingly, in mouse models, schistosome eggs or egg-derived antigens are considered potent inducers of a Th2-type immune response. A robust Th2 response has been shown to play a significant role in the development of granulomas around deposited eggs [57,69]. Numerous investigations have reported significant variations in histopathological findings between mice receiving treatment with natural products and those that did not, as evidenced by marked decreases in hepatic granulomas, fibrosis, and pro-inflammatory cytokines [53,55,66,67]. The recently evaluated compound plumbagin (5-hydroxy-3-methyl-1,4-naphthoquinone), derived from walnut trees, has been demonstrated to alleviate schistosome-induced hepatosplenomegaly, as well as to reduce hepatic granuloma and liver collagen content by 62.5% and 35.3%, respectively. Plumbagin exhibited immunomodulatory properties, as evidenced by increased levels of IL-10, while levels of IL-4, IL-13, IL-17, IL-37, IFN-γ, TGF-β, and TNF-α decreased [66]. Interestingly, a similar immunomodulatory profile was observed from another phenolic compound derived from the same source, Juglone (5-hydroxy-1,4-naphthoquinone) [67].
Studies conducted by El-Aal et al. [55] demonstrated that treatment with Paeoniflorin, a potential anti-schistosomal therapy, led to higher serum levels of TNF-α compared to healthy controls, infected controls, and Praziquantel-treated mice. The literature contains conflicting information regarding the complex role of TNF-α in schistosomiasis. While this cytokine can initiate apoptosis and provoke anti-fibrogenic effects, it has also been associated with granuloma formation and a fibrotic tissue development [50,55,56].
The oral administration of different natural compounds at doses ranging between 1.5 and 400 mg/kg demonstrated variable worm and egg burden reduction (31.8 to 100%) in infected mice [44,53,54,60,63,64]. In addition, Carvalho et al. [65] demonstrated the effective antischistosomal activity of asiaticoside (400 mg/kg) against Schistosoma spp. by inhibiting SmNTPDases—enzymes found in the worm tegument—which resulted in significantly reduced worm burden.
The findings in the reviewed studies indicate the natural compounds used for treatment via intraperitoneal route were all of the phenol class, or its derivatives [43,50,66,67]. For example, licochalcone A exhibits significant potential as a therapeutic drug. However, recent pharmacokinetic studies have indicated that its oral bioavailability is limited due to poor absorption and inactivation. Thus, alternative routes of administration, such as intraperitoneal or conjugation with nanoparticles, may be required to enhance therapeutic efficacy [62].
Several studies have shown female worms to be more susceptible to antischistosomal drugs than males [43,53,67]. In this scenario, Juglone has been proven to exert efficacious schistomicidal activity. Naphthoquinones react with the thiol groups of S. mansoni parasite proteins and inhibit enzymes essential for parasite survival [67]. Juglone was shown to significantly reduce the burden of both male and female worms by 63.12% and 52.1%, respectively. Notably, this compound demonstrated activity against both male and female worms, unlike other compounds that exhibit selective activity against one sex over another.

2.3. Essential Oils and Their Components in Use against Schistosoma Mansoni

Essential oils are characterized by a mixture of volatile and hydrophobic secondary metabolites. These oils constitute one of the principal fractions of chemical substances found in plants, presenting marked odors and being composed primarily of terpenoids and phenylpropanoids [70]. Essential oils have been evaluated for their anti-schistosomal potential at different stages of the parasite life cycles.
In a molluscicidal evaluation, Eucalyptus essential oils demonstrated bioactivity against B. glabrata eggs [71]. Essential oil extracted from Eryngium triquetrum, which contains aliphatic polyacetylene, also was found to be toxic to infected snails, exerting moderate effects on B. glabrata embryos in terms of inhibited egg hatching and snail development [72]. Among the compounds evaluated, the activity of terpene compounds found in some oils was described. The commercially available monoterpenes thymol and α-pinene demonstrated activity against B. glabrata snails, inducing mortality in a concentration-dependent manner, as well as inhibiting the enzymatic activity of acetylcholinesterase (AChE) extracted from snails [73]. In the context of biotechnology applications, nanoemulsions as a vehicle of the essential oil of Xylopia ochrantha (main compounds: bicyclogermacrene and germacrene D) caused between 50 to 100% mortality in B. tenagophila, B. straminea, and B. glabrata juveniles and adults after 48 h, and inhibited the development of eggs deposited by treated snails [74].
The chemoprophylactic action of Pterodon pubescens essential oil as an additive in different soap formulations was studied. Following the application of solutions containing different soap concentrations to the tails of mice, the animals were infected immediately, or 24 h later, with S. mansoni via caudal immersion. After 45 days of infection, different levels of protection were observed, ranging from 29 to 100% [75].
Cercaricidal activity was described for essential oil from the fresh aerial part of Apium graveolens var. secalinum (alpha- and beta-pinene, myrcene, limonene, cis-beta-ocimene, gamma-terpinene, cis-allo-ocimene, trans-farnesene, humulene, apiol, beta-selinene, senkyunolide, and neocnidilide) and in essential oils of Eucalyptus spp. (E. cloeziana, E. deanei, E. exserta, E. maculata, E. punctate, and E. resinifera) [71,76]. In studies using cedar oil, the authors attributed optimal cercaricidal activity to the penetration phase of cercariae, in which disruption of the cercarial glycocalyx alters the physiological processes related to osmoregulation, and may increase the absorption of toxic substances [77].
In in vitro studies involving S. mansoni, essential oil from the leaves of B. dracunculifolia, constituted mainly of oxygenated sesquiterpenes, such as (E)-nerolidol (33.51%) and spathulenol (16.24%), demonstrated high activity in a schistosomicidal assay, leading to the death of cultured pairs of adult worms [78]. Essential oil from Plectranthus neochilus, consisting of b-caryophyllene (1; 28.23%), a-thujene (2; 12.22%), a-pinene (3; 12.63%), b-pinene (4; 6.19%), germacrene D (5; 5.36%), and caryophyllene oxide (6; 5.37%), was considered active, but less effective than chemotherapy treatment in terms of worm pair separation, mortality, decreased motor activity, and tegumentary changes. However, this oil was associated with a dose-dependent reduction in both number and percentage of S. mansoni eggs [79]. Essential oil from Ageratum conyzoides L., whose main constituents are precocele I (74.30%) and (E)-caryophyllene (14.23%), was also considered less active than PZQ in the in vitro treatment of worms, with a dose-dependent reduction observed in the number of S. mansoni eggs [80]. Essential oil obtained from Tetradenia riparia leaves also reduced motility and decreased the percentage of developed eggs [81]. When S. mansoni worms were incubated with Mentha × villosa essential oil and its individual constituents (rotundifolone (70.96%), limonene (8.75%), transcaryophyllene (1.46%), and β-pinene (0.81%)), no anti-schistosomicidal activities were observed for transcaryophyllene or β-pinene. However, the use of this essential oil and rotundifolone or limonene did result in decreased adult worm motility and increased mortality [82].
In vitro assays involving Baccharis trimera demonstrated motility loss and death in S. mansoni within 30 h after exposure. Morphological alterations in the tegument were described in male worms, indicating desquamation on the surface of the tegument, as well as the destruction of tubercles and spines, resulting in smooth body surface areas. This essential oil also caused integumentary destruction in female worms, in addition to the destruction of the oral and acetabular cups [83]. Ultrastructural worm evaluation revealed bubble lesions, loss of tubercles in some regions of the ventral portion, tegument alterations, vacuoles in the region of the syncytial matrix, and glycogen granules near the muscle fibers [84]. Essential oil from Foeniculum vulgare MILL, whose main constituents are (E)-anethole (69.8%) and limonene (22.5%), exerted inhibitory effects on S. mansoni egg development, with less effective results than the positive control (PZQ) in terms of mated pair separation, mortality, and decreased motor activity [85]. Essential oils obtained from Citrus limonia leaves (Limonene-29.9%, β-pinene-12.0%, sabinense-9.0%, citronellal-9.0%, and citronellol-5.8%) and C. reticulata fruit peels (limonene-26.5%, γ-terpinene-17.2%, linalool-11.1%, octanal-8.0%, myrcene-6.2%, and capraldehyde-3.9%) exhibited moderate in vitro schistosomicidal activity against adult S. mansoni worms [86]. Essential oil from Dysphania ambrosioides (L.) (main constituents: cis-piperitone oxide monoterpenes-35.2%, p-cymene-14.5%, isoascaridol-14.1%, and a-terpinene-11.6%) showed in vitro parasite mortality in 100% of adult worm pairs after 24 h of treatment [87].
Although several in vitro studies have demonstrated the effects of different essential oils on S. mansoni, few in vivo studies have been conducted. Treatment with essential oil from fresh Melaleuca armillaris leaves (main constituents: 1,8-cineol-33.93%, terpinen-4-ol-18.79%, limonene-10, 37%, and B-pinene-6.59%) administered in mice twice a week for six weeks (150 mg/kg, orally), from the second week post-infection, significantly improved levels of glutathione and malondialdehyde and raised levels of vitamins C and E [88]. Matos-Rocha, 2020, [61] treated infected mice with Mentha × villosa essential oil (200 mg/kg) and rotundifolone (141.9 mg/kg) for five consecutive days, observing respective reductions of 72.44% and 74.48% in recovered S. mansoni after treatment.

3. Useful Tools for the Screening of New Drugs in Schistosomiasis

To reduce the time and costs associated with conventional methods of determining anti-Schistosoma activity in new drugs, which normally include laborious manual experiments based on phenotypic analysis by microscopy, several computational and experimental tools have been developed to enhance the search for new compounds [89,90,91]. These tools can not only aid in the identification of new substances with antischistosomal potential, whether of natural, synthetic, or semi-synthetic origin, but also aim to determine drug-likeness and guide proximal screening steps with regard to biological activity.
The approaches utilized for the selection and design of new compounds include structure-based drug design, in which a potential molecular target is characterized, as well as its participation in some metabolic event related to disease. In this type of approach, the use of three-dimensional (3D) molecular target structure, whether validated or putative, is essential in conducting protein–ligand interaction studies, as well as evaluating the forces involved in these interactions. This type of approach permits enhanced in silico virtual screening (VS) capability [92]. By contrast, drug design based on ligands does not necessarily depend on the structure of the potential target, but rather estimates parameters of the ligands themselves, such as structure, activity, and other important properties related to biological activity and drug-likeness. The most common methods used to perform these estimations are QSAR (quantitative structure–activity relationship) and QSPR (quantitative structure–property relationship) [93].
Computational methods employ algorithms and simulations that help predict physical–chemical characteristics and the potential for interaction between molecules through molecular docking, molecular dynamics, and molecular mechanics. Several studies have used these types of tools in the search for new compounds with activity against schistosomiasis, such as CADD (Computer-Aided Drug Design) and QSAR modeling. CADD enables the performance of virtual compound library screening and can simulate chemical modifications in compounds with already-known activity, aiming to improve physicochemical characteristics and enhance interaction with specific target ligands. Computational techniques for drug screening also include in silico analysis and methods for large-scale experimental data analysis, such as those evaluated by High Throughput Screening (HTS) [91].
In this context, databases of compound structures and molecular models of the main targets of the S. mansoni parasite have aided in the process of developing robust in silico tools that provide refined and reliable 3D structures. Moreover, genomic and protein databases play a very important role by providing sequences to obtain high-quality molecular models for investigation using in silico tools. The genomes of S. mansoni, as well as related species S. japonicum and S. haematobium, were first published in 2009, followed by updated versions [94,95], and can currently be freely accessed via the Wormbase ParaSite database (https://parasite.wormbase.org/; accessed on 15 May 2023). Recently, the availability of whole-genome sequencing and transcriptomic analysis has demonstrated the importance of a deeper understanding of this parasite’s gene expression. The integration of data can not only advance the development of novel schistosomiasis control strategies, especially regarding new drug discovery, but also aid in many diverse aspects of investigation, including variations in parasites that may reflect treatment response [96,97].
Concerning molecular targets, a variety of proteins have been identified as potential target molecules for the action of drugs and inhibitors. The protein data bank (PDB) (https://www.rcsb.org; accessed on 15 May 2023) is a repository containing experimentally determined 3D protein structures and protein models defined by computers, both of which are employed in CADD approaches. This database contains over 140,000 structures of S. mansoni proteins and protein domains, alone or with ligand complexes. A summary of the main potential drug targets of Schistosoma are presented in a study by Cheuka (2022) [98] along with information on function, method of identification/validation, examples of inhibitors or antagonists, and relevant phenotypic effects on schistosomes. Of these, some molecules are widely known and investigated, such as thioredoxin glutathione reductase (TGR), glutathione-S-transferase (GST), histone deacetylase (HDAC), and 20S proteasome, while others have been more recently identified and require further validation [98].
Currently, the use of artificial intelligence and machine learning have been integrated into tools that both seek to predict the diagnosis of disease [99] and the generation of three-dimensional models, in an attempt to reverse certain barriers, such as the need for crystallographic structures prior to the determination of reliable structural models. In this context, the launch of the AlphaFold tool [100] represents a revolution in obtaining structural models, as this prediction system employs artificial neural networks to accurately and rapidly predict 3D protein structures from primary amino acid sequences, without necessarily utilizing a previously developed model. Applications in schistosomiasis studies include obtaining new protein models, some of which can be found in the AlphaFold Protein Structure Database (AlphaFold DB) (https://www.alphafold.ebi.ac.uk/; accessed on 14 May 2023), which currently contains more than 15,000 structures for S. mansoni, including the main drug targets SmTGR and SmGST.
Several studies have used these tools to facilitate the choice of targets and/or optimal drug candidates, in addition to algorithms that promote a more efficient understanding of data produced on a large scale, as in HTS phenotypic analysis [91].
Through the application of CADD using the Molecular Operating Environment (MOE) tool to optimize compound structures and evaluate drug-likeness, 27 compounds and derivatives of African medicinal plants previously reported to present anti-Schistosoma effects in vitro or in vivo were evaluated. Computational analysis reduced screening candidates to just four molecules with potential activity on Schistosoma molecular targets, including TGR, GST, HDAC, and arginase [101].
The combination of structure- and ligand-based screening methods was also applied in the virtual screening of 1000 alkaloid structures isolated from plants of the Menispermaceae and Apocynaceae families to detect potential action against S. mansoni. This study employed QSAR approaches based on chemoinformatic tools available at the website Openmolecules.org, such as DataWarrior, to identify two alkaloids for use as a starting point for the development of new chemical compounds with anti-Schistosoma activity [102].
With regard to metabolic targets, QSAR-based virtual screening of S. mansoni thioredoxin glutathione reductase (SmTGR) inhibitors was performed using high content screening (HCS) in an effort to discover novel antischistosomal agents. QSAR models aimed at inhibiting SmTGR were applied to three subsets from the ChemBridge library (∼150,000 compounds), which selected 29 compounds for further testing via two HCS platforms based on image analysis of assay plates. Among these, 2-[2-(3-methyl-4-nitro-5-isoxazolyl)vinyl]pyridine and 2-(benzylsulfonyl)-1,3-benzothiazole, two compounds representing the new chemical scaffolds, were found to exert activity against schistosomula and adult worms at low micromolar concentrations, thus constituting promising antischistosomal agents [103]. A combination of computational techniques that included molecular docking studies also allowed for the evaluation of approximately 1000 insect-derived compounds with demonstrated inhibitory activity against SmTGR [103]. The applied approach included both the creation of a database of molecules derived from insects and the use of the PLIP (protein–ligand interaction profiler) tool for virtual screening and selection of the best candidates to conduct experimental testing.
Another example of combining CADD methods for the virtual screening of anti-S. mansoni drugs based on molecular docking is evidenced in a study by Moreira et al., in which a panel of 85,000 molecules from the Managed Chemical Compounds Collection (MCCC) of the University of Nottingham (UK) were investigated against five protein kinases (JNK, p38, ERK1, ERK2, and FES). Computational analysis narrowed the initial number of molecules down to 169, which were predicted to bind to SmERK1, SmERK2, SmFES, SmJNK, and/or Smp38, and were thus selected for testing in in vitro screening assays using schistosomula and adult worms. This combination of in silico and in vitro assays helped optimize the search for the most promising compounds, leading to a total of 89 molecules that were considered active by experimental assays [104], with 17 having suitable drug-likeness parameters. One of the experimental approaches aimed at detecting movement in adult worms was the WormAssay, a high-throughput screening motility assay that simultaneously performs parallel analysis on all wells of an entire plate [105]. WormAssay is another example of a useful, low-cost computer-aided tool that can be applied in the search for molecules against schistosomes.
In the search for inhibitors of venus kinase receptors (VKR), important to schistosome growth and egg deposition, 645 molecules from GlaxoSmithKline (GSK) set 2 were screened against one of the target proteins of S. mansoni (SmVKR2). This strategy combined the use of an initial in vitro experimental screening approach that applied the surface plasmon resonance (SPR) technique to determine molecule binding constants, with just 12 demonstrating molecular interactions on a micromolar level. These twelve molecules were then tested against S. mansoni ex vivo, resulting in the identification of four compounds with antiparasitic activity under testing including the WormAssay for phenotypic determination. Furthermore, the crystal structure of the kinase domain of SmVKR2 obtained by the authors improved in silico docking, thus paving the way to identify more potent inhibitors against the VKR2 receptor in the future [106].
Targeting potential epigenomic effects on the parasite, a structure-based virtual screening approach was applied in the search for histone deacetylase 8 (SmHDAC8) inhibitors using molecular docking of a compound library containing 550,000 molecules (the Interbioscreen database) against the crystallized structure of SmHDAC8 deposited in the PDB. The Glide docking program identified eight novel N-(2,5-dioxopyrrolidin-3-yl)-n-alkylhydroxamate derivatives, which were found to be active in the low micromolar range against smHDAC8 by utilizing an established in vitro assay for protein–ligand interaction and apoptosis induction [107].

4. Strategies Employing Functional Genomics Approaches and New Perspectives

Modern methods and technologies that combine functional genomic approaches, including genomics, transcriptomics, proteomics and epigenetics of S. mansoni, have shown the importance of achieving a deeper understanding of this parasite’s gene expression and the integration of data to advance the development of schistosomiasis control strategies. This is relevant not only with respect to the new drug discovery, but also in many diverse aspects, including populational variations that may reflect on the response to treatment, as well as mechanisms involved in the development of resistance to chemotherapy [96,97].
The genome of S. mansoni, as well as related species S. japonicum and S. haematobium, was first published in 2009, followed by updated revisions [94,95] which are freely available on the WormBase ParaSite database (https://parasite.wormbase.org/; accessed on 14 June 2023). Genomic data from publicly available databases serve as resources for establishing relationships between drugs and targets, allowing for the identification of compounds from searches based on sequence homology and functional motifs. Neves and colleagues demonstrated the use of this type of approach in their search for drug repositioning for schistosomiasis [102]. Whole-genome sequencing has also been applied with the aim of elucidating the impact of treatment on the parasite genome. For example, the action of PZQ was evaluated on natural populations of S. mansoni in areas where mass drug administration strategies had been conducted [108,109].
Data available from proteomics studies permit greater knowledge surrounding the profile of differentially expressed proteins under a variety of conditions to which the parasite is exposed. A comparative study of the excretory/secretory proteome of adult male and female worms identified approximately 1000 proteins, of which 370 and 140 were secreted solely or abundantly by males and females, respectively. The use of functional genomic analysis tools served to indicate which classes of proteins were more related to secretion by males than females, providing valuable information on host–parasite interplay and male–female interaction [110].
The response to treatment on the parasite’s proteome is also a relevant topic of investigation. For example, laboratory resistance selection of S. mansoni isolates through exposure to PZQ enabled the evaluation of impact on protein expression profile in worms exhibiting reduced sensitivity to PZQ in comparison to a susceptible population. This resulted in the identification of two proteins, Ca2+-ATPase and HSP70, that revealed differentiated expression under diverse analyzed conditions [111]. PZQ-resistant isolates were also compared regarding global changes in gene expression, with emphasis placed on determining the differential proteome between male and female worms. This approach enabled the identification of 60 differentially expressed proteins between exposed and non-exposed populations, with some proteins detected exclusively in females, whose susceptibility to PZQ was reduced in relation to male worms [112]. This type of approach greatly enhances the understanding of potential resistance mechanisms and also aids in establishing potential correlations with the drug’s mode of action.
In a similar context, many studies have also evaluated the profile of differentially expressed genes through RNA-seq, which has proven to be a very robust tool in identifying altered mRNA levels under both natural and experimental conditions [113,114]. More recently, transcriptomic single cell analysis has shown great potential in answering questions regarding parasite development, as well as its interaction with the environment and the host, unraveling possible heterogenous responses and transcriptomic dynamics [115,116].
Studies employing a combination of strategies, including bioinformatics tools, cheminformatics, and functional genomics, have been shown to efficiently select molecular targets involved in epigenetic regulatory processes, as reported by Padalino et al. [117], who utilized sequence homology, phylogenetic analysis, and a refined search for functional motifs, which, following experimental validation, permitted the reclassification of two components of the histone methylation machinery. In addition, posterior investigation of the potential of inhibition of these targets demonstrated the possibility of exploring the epigenetic pathway towards the development of next-generation drugs targeting schistosome epigenetic pathway components [117].
Thus, the use and integration of “omics” technologies has provided data deemed fundamental to defining new perspectives on the identification and application of natural products with anti-schistosomal properties.

5. Concluding Remarks

In conclusion, natural compounds have demonstrated significant potential as antischistosomal agents. Both in vitro and in vivo studies have evidenced the effectiveness of natural compounds against schistosomes under various approaches, including prophylactic interventions and adult parasite, cercariae, and schistosomula killing, as well as suppressive strategies that inhibit worm egg-laying. However, specific mechanisms of action, toxicological testing, and in vivo activities require better characterization in further investigations to allow transposing the use of natural products in clinical studies involving subjects with schistosomiasis. Overall, natural products have provided a valuable source of potential antischistosomal agents that may aid in the development of new treatments for schistosomiasis, a disease that continues to cause significant morbidity and mortality in endemic areas.

Author Contributions

C.M.A.: literature review, corrections, and manuscript writing. C.S.M.: manuscript review, corrections, and editing. J.W.d.S.: literature review and manuscript writing. D.M.N.M.: literature review and manuscript writing. S.A.d.O.: manuscript review and editing. C.J.d.C.: literature review and manuscript writing. E.d.S.S.: manuscript review and editing. M.B.P.S.: manuscript review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank Andris K. Walter for critical analysis, English language revision, and manuscript copyediting assistance.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Not applicable.

References

  1. Lo, N.C.; Bezerra, F.S.M.; Colley, D.G.; Fleming, F.M.; Homeida, M.; Kabatereine, M.; Kabole, F.M.; King, C.H.; Mafe, M.A.; Midzi, M.; et al. Review of 2022 WHO guidelines on the control and elimination of schistosomiasis. Lancet Infect Dis. 2022, 22, e327–e335. [Google Scholar] [CrossRef]
  2. McManus, D.P.; Bergquist, R.; Cai, P.; Ranasinghe, S.; Tebeje, B.M.; You, H. Schistosomiasis—From immunopathology to vacines. Semin. Immunopathol. 2020, 42, 355–371. [Google Scholar] [CrossRef]
  3. LoVerde, P.T. Schistosomiasis. Adv. Exp. Med. Biol. 2019, 1154, 45–70. [Google Scholar] [CrossRef]
  4. Wu, G.Y.; Halim, M.H. Schistosomiasis: Progress and problems. World J. Gastroenterol. 2000, 6, 12–19. [Google Scholar] [CrossRef]
  5. Gryseels, B.; Polman, K.; Clerinx, J.; Kestens, L. Human schistosomiasis. Lancet 2006, 368, 1106–1118. [Google Scholar] [CrossRef]
  6. Whitfield, P.J.; Bartlett, A.; Khammo, N.; Clothier, R.H. Age-dependent survival and infectivity of Schistosoma mansoni cercariae. Parasitology 2003, 127 Pt 1, 29–35. [Google Scholar] [CrossRef] [PubMed]
  7. Neves, B.J.; Andrade, C.H.; Cravo, P.V. Natural products as leads in schistosome drug discovery. Molecules 2015, 20, 1872–1903. [Google Scholar] [CrossRef] [PubMed]
  8. McManus, D.P.; Dunne, D.W.; Sacko, M.; Utzinger, J.; Vennervald, B.J.; Zhou, X.N. Schistosomiasis. Nat. Rev. Dis. Primers 2018, 4, 13. [Google Scholar] [CrossRef]
  9. Siqueira, L.D.P.; Fontes, D.A.F.; Aguilera, C.S.B.; Timóteo, T.R.R.; Ângelos, M.A.; Silva, L.C.P.B.B.; de Melo, C.G.; Rolim, L.A.; da Silva, R.M.F.; Neto, P.J.R. Schistosomiasis: Drugs used and treatment strategies. Acta Trop. 2017, 176, 179–187. [Google Scholar] [CrossRef]
  10. Colley, D.G.; Bustinduy, A.L.; Secor, W.E.; King, C.H. Human schistosomiasis. Lancet 2014, 383, 2253–2264. [Google Scholar] [CrossRef] [PubMed]
  11. Ross, A.G.; Bartley, P.B.; Sleigh, A.C.; Olds, G.R.; Li, Y.; Williams, G.M.; McManus, D.P. Schistosomiasis. N. Engl. J. Med. 2002, 346, 1212–1220. [Google Scholar] [CrossRef] [PubMed]
  12. Thétiot-Laurent, S.A.; Boissier, J.; Robert, A.; Meunier, B. Schistosomiasis chemotherapy. Angew. Chem. Int. Ed. Engl. 2013, 52, 7936–7956. [Google Scholar] [CrossRef] [PubMed]
  13. World Health Organization. Available online: http://www.who.int/schistosomiasis/strategy/en/ (accessed on 15 January 2023).
  14. Tomiotto-Pellissier, F.; Miranda-Sapla, M.M.; Machado, L.F.; Bortoleti, B.T.S.; Sahd, C.S.; Chagas, A.F.; Assolini, J.P.; Oliveira, F.J.A.; Pavanelli, W.R.; Conchon-Costa, I.; et al. Nanotechnology as a potential therapeutic alternative for schistosomiasis. Acta Trop. 2017, 174, 64–71. [Google Scholar] [CrossRef]
  15. Cioli, D. Chemotherapy of Schistosomiasis: An Update. Parasitol. Today 1998, 14, 418–422. [Google Scholar] [CrossRef] [PubMed]
  16. Caffrey, C.R. Schistosomiasis and its treatment. Future Med. Chem. 2015, 7, 675–676. [Google Scholar] [CrossRef]
  17. Mäder, P.; Rennar, G.A.; Ventura, A.M.P.; Grevelding, C.G.; Schlitzer, M. Chemotherapy for Fighting Schistosomiasis: Past, Present and Future. Chem. Med. Chem. 2018, 13, 2374–2389. [Google Scholar] [CrossRef]
  18. Adegboye, O.; Field, M.A.; Kupz, A.; Pai, S.; Sharma, D.; Smout, M.J.; Wangchuk, P.; Wong, Y.; Loiseau, C. Natural-Product-Based Solutions for Tropical Infectious Diseases. Clin. Microbiol. Rev. 2021, 34, e0034820. [Google Scholar] [CrossRef]
  19. Eze, A.A.; Ogugofor, M.O.; Ossai, E.C. Plant-Derived Compounds for the Treatment of Schistosomiasis: Improving Efficacy Via Nano-Drug Delivery. Niger. J. Clin. Pract. 2022, 25, 747–764. [Google Scholar] [CrossRef]
  20. Mushtaq, S.; Abbasi, B.H.; Uzair, B.; Abbasi, R. Natural products as reservoirs of novel therapeutic agents. EXCLI J. 2018, 17, 420–451. [Google Scholar] [CrossRef]
  21. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed]
  22. Cunha, N.L.; Uchôa, C.J.D.M.; Cintra, L.S.; Souza, H.C.D.; Peixoto, J.A.; Silva, C.P.; Magalhães, L.G.; Gimenez, V.M.M.; Groppo, M.; Rodrigues, V.; et al. In vitro Schistosomicidal Activity of Some Brazilian Cerrado Species and Their Isolated Compounds. Evid.-Based Complement. Altern. Med. 2012, 2012, 173614. [Google Scholar] [CrossRef] [PubMed]
  23. Moraes, J.; Nascimento, C.; Yamaguchi, L.F.; Kato, M.J.; Nakano, E. Schistosoma Mansoni: In vitro Schistosomicidal Activity and Tegumental Alterations Induced by Piplartine on Schistosomula. Exp. Parasitol. 2012, 132, 222–227. [Google Scholar] [CrossRef]
  24. Matos, J.L.; da Silva, K.R.; de Lima Paula, L.A.; Cunha, W.R.; Ramos, S.B.; Rodrigues, V.; Cabral, F.J.; Magalhães, L.G. Molluscicidal and Cercaricidal Activities of Curcumin on Biomphalaria glabrata and Schistosoma mansoni Cercariae. Pest. Manag. Sci. 2020, 76, 1228–1234. [Google Scholar] [CrossRef] [PubMed]
  25. Islam, M.T.; Martorell, M.; Salehi, B.; Setzer, W.N.; Sharifi-Rad, J. Anti-Schistosoma mansoni effects of essential oils and their components. Phytother. Res. 2020, 34, 1761–1769. [Google Scholar] [CrossRef] [PubMed]
  26. Silva, M.P.N.; Oliveira, G.S.L.; de Carvalho, R.B.F.; de Sousa, D.P.; Freitas, R.M.; Pinto, P.L.S.; de Moraes, J. Antischistosomal Activity of the Terpene Nerolidol. Molecules 2014, 19, 3793–3803. [Google Scholar] [CrossRef] [PubMed]
  27. Ramalhete, C.; Magalhães, L.; Rodrigues, V.; Mulhovo, S.; Da Silva Filho, A.A.; Ferreira, M.J.U. In vitro Schistosomicidal Activity of Balsaminol F and Karavilagenin, C. Planta Med. 2012, 78, 1912–1917. [Google Scholar] [CrossRef] [PubMed]
  28. Silva, I.P.; Brissow, E.; Kellner Filho, L.C.; Senabio, J.; de Siqueira, K.A.; Vandresen Filho, S.; Damasceno, J.L.; Mendes, S.A.; Tavares, D.C.; Magalhães, L.G.; et al. Bioactive Compounds of Aspergillus Terreus—F7, an Endophytic Fungus from Hyptis suaveolens (L.) Poit. World J. Microbiol. Biotechnol. 2017, 33, 62. [Google Scholar] [CrossRef] [PubMed]
  29. Moraes, J.; Nascimento, C.; Miura, L.M.C.V.; Leite, J.R.S.A.; Nakano, E.; Kawano, T. Evaluation of the in vitro Activity of Dermaseptin 01, a Cationic Antimicrobial Peptide, against Schistosoma mansoni. Chem. Biodivers. 2011, 8, 548–558. [Google Scholar] [CrossRef] [PubMed]
  30. Moraes, J.; Almeida, A.A.C.; Brito, M.R.M.; Marques, T.H.C.; Lima, T.C.; De Sousa, D.P.; Nakano, E.; Mendonça, R.Z.; Freitas, R.M. Anthelmintic Activity of the Natural Compound (+)-Limonene Epoxide against Schistosoma mansoni. Planta Med. 2013, 79, 253–258. [Google Scholar] [CrossRef]
  31. Carrara, V.S.; Vieira, S.C.H.; De Paula, R.G.; Rodrigues, V.; Magalhães, L.G.; Cortez, D.A.G.; Da Silva Filho, A.A. In vitro Schistosomicidal Effects of Aqueous and Dichloromethane Fractions from Leaves and Stems of Piper species and the Isolation of an Active Amide from P. Amalago L. (Piperaceae). J. Helminthol. 2014, 88, 321–326. [Google Scholar] [CrossRef]
  32. Santos, A.F.; Fonseca, S.A.; César, F.A.; De Azevedo Albuquerque, M.C.P.; Santana, J.V.; Santana, A.E.G. A Penta-Substituted Pyridine Alkaloid from the Rhizome of Jatropha elliptica (Pohl) Muell. Arg. Is Active against Schistosoma mansoni and Biomphalaria glabrata. Parasitol. Res. 2014, 113, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
  33. Aleixo de Carvalho, L.S.; Geraldo, R.B.; de Moraes, J.; Silva Pinto, P.L.; de Faria Pinto, P.; Pereira, O.d.S.; Da Silva Filho, A.A. Schistosomicidal Activity and Docking of Schistosoma mansoni ATPDase 1 with Licoflavone B Isolated from Glycyrrhiza inflata (Fabaceae). Exp. Parasitol. 2015, 159, 207–214. [Google Scholar] [CrossRef] [PubMed]
  34. Alvarenga, T.A.; De Oliveira, P.F.; De Souza, J.M.; Tavares, D.C.; Andrade, E.; Silva, M.L.; Cunha, W.R.; Groppo, M.; Januário, A.H.; Magalhães, L.G.; et al. Schistosomicidal Activity of Alkyl-Phenols from the Cashew Anacardium occidentale against Schistosoma mansoni Adult Worms. J. Agric. Food Chem. 2016, 64, 8821–8827. [Google Scholar] [CrossRef] [PubMed]
  35. Eraky, M.A.; Aly, N.S.M.; Selem, R.F.; El-Kholy, A.A.E.M.; Rashed, G.A.E.R. In vitro Schistosomicidal Activity of Phytol and Tegumental Alterations Induced in Juvenile and Adult Stages of Schistosoma haematobium. Korean J. Parasitol. 2016, 54, 477–484. [Google Scholar] [CrossRef]
  36. Pereira, V.R.D.; Junior, I.J.A.; da Silveira, L.S.; Geraldo, R.B.; Pinto, P.d.F.; Teixeira, F.S.; Salvadori, M.C.; Silva, M.P.; Alves, L.A.; Capriles, P.V.S.Z.; et al. In vitro and in vivo Antischistosomal Activities of Chalcones. Chem. Biodivers. 2018, 15, e1800398. [Google Scholar] [CrossRef]
  37. Parreira, R.L.T.; Costa, E.S.; Heleno, V.C.G.; Magalhães, L.G.; Souza, J.M.; Pauletti, P.M.; Cunha, W.R.; Januário, A.H.; Símaro, G.V.; Bastos, J.K.; et al. Evaluation of Lignans from Piper Cubeba against Schistosoma mansoni Adult Worms: A Combined Experimental and Theoretical Study. Chem. Biodivers. 2019, 16, e1800305. [Google Scholar] [CrossRef] [PubMed]
  38. Dube, M.; Saoud, M.; Rennert, R.; Fotso, G.W.; Andrae-Marobela, K.; Imming, P.; Häberli, C.; Keiser, J.; Arnold, N. Anthelmintic Activity and Cytotoxic Effects of Compounds Isolated from the Fruits of Ozoroa insignis Del. (Anacardiaceae). Biomolecules 2021, 11, 1893. [Google Scholar] [CrossRef] [PubMed]
  39. Sirak, B.; Asres, K.; Hailu, A.; Dube, M.; Arnold, N.; Häberli, C.; Keiser, J.; Imming, P. In vitro Antileishmanial and Antischistosomal Activities of Anemonin Isolated from the Fresh Leaves of Ranunculus multifidus Forsk. Molecules 2021, 26, 7473. [Google Scholar] [CrossRef] [PubMed]
  40. Tirichen, H.; Yaigoub, H.; Xu, W.; Wu, C.; Li, R.; Li, Y. Mitochondrial Reactive Oxygen Species and Their Contribution in Chronic Kidney Disease Progression through Oxidative Stress. Front. Physiol. 2021, 12, 627837. [Google Scholar] [CrossRef]
  41. Brand, M.D. Mitochondrial generation of superoxide and hydrogen peroxide as the source of mitochondrial redox signaling. Free. Radic. Biol. Med. 2016, 100, 14–31. [Google Scholar] [CrossRef] [PubMed]
  42. Guerra-Castellano, A.; Díaz-Quintana, A.; Pérez-Mejías, G.; Elena-Real, C.A.; González-Arzola, K.; García-Mauriño, S.M.; De la Rosa, M.A.; Díaz-Moreno, I. Oxidative stress is tightly regulated by cytochrome c phosphorylation and respirasome factors in mitochondria. Biophys. Comput. Biol. 2018, 115, 7955–7960. [Google Scholar] [CrossRef]
  43. Allam, G.; Abuelsaad, A.S.A. In vitro and in vivo Effects of Hesperidin Treatment on Adult Worms of Schistosoma mansoni. J. Helminthol. 2014, 88, 362–370. [Google Scholar] [CrossRef]
  44. Moraes, J.; de Oliveira, R.N.; Costa, J.P.; Junior, A.L.G.; de Sousa, D.P.; Freitas, R.M.; Allegretti, S.M.; Pinto, P.L.S. Phytol, a Diterpene Alcohol from Chlorophyll, as a Drug against Neglected Tropical Disease Schistosomiasis Mansoni. PLoS Negl. Trop. Dis. 2014, 8, 51. [Google Scholar] [CrossRef] [PubMed]
  45. Reimer, A.; Blohm, A.; Quack, T.; Grevelding, C.G.; Kozjak-Pavlovic, V.; Rudel, T.; Hentschel, U.; Abdelmohsen, U.R. Inhibitory Activities of the Marine Streptomycete-Derived Compound SF2446A2 against Chlamydia Trachomatis and Schistosoma mansoni. J. Antibiot. 2015, 68, 674–679. [Google Scholar] [CrossRef] [PubMed]
  46. Mafud, A.C.; Silva, M.P.N.; Monteiro, D.C.; Oliveira, M.F.; Resende, J.G.; Coelho, M.L.; De Sousa, D.P.; Mendonça, R.Z.; Pinto, P.L.S.; Freitas, R.M.; et al. Structural Parameters, Molecular Properties, and Biological Evaluation of Some Terpenes Targeting Schistosoma mansoni Parasite. Chem. Biol. Interact. 2016, 244, 129–139. [Google Scholar] [CrossRef]
  47. Martins, M.C.B.; Silva, M.C.; Silva, H.A.M.F.; Silva, L.R.S.; De Azevedo Albuquerque, M.C.P.; Aires, A.L.; Da Silva Falcão, E.P.; Pereira, E.C.; De Melo, A.M.M.A.; De Silva, N.H. Barbatic Acid Offers a New Possibility for Control of Biomphalaria glabrata and Schistosomiasis. Molecules 2017, 22, 568. [Google Scholar] [CrossRef] [PubMed]
  48. Souza, R.L.; Gonçalves, U.O.; Badoco, F.R.; de Souza Galvão, L.; Santos, R.A.d.; de Carvalho, P.H.D.; de Carvalho, L.S.A.; da Silva Filho, A.A.; Veneziani, R.C.S.; Rodrigues, V.; et al. Licochalcone A Induces Morphological and Biochemical Alterations in Schistosoma mansoni Adult Worms. Biomed. Pharmacother. 2017, 96, 64–71. [Google Scholar] [CrossRef] [PubMed]
  49. Lago, E.M.; Xavier, R.P.; Teixeira, T.R.; Silva, L.M.; da Silva Filho, A.A.; de Moraes, J. Antischistosomal Agents: State of art and perspectives. Future Med. Chem. 2018, 10, 89–120. [Google Scholar] [CrossRef]
  50. Allam, G. Immunomodulatory Effects of Curcumin Treatment on Murine Schistosomiasis Mansoni. Immunobiology 2009, 214, 712–727. [Google Scholar] [CrossRef] [PubMed]
  51. El-Agamy, D.S.; Shebl, A.M.; Said, S.A. Prevention and Treatment of Schistosoma mansoni-Induced Liver Fibrosis in Mice. Inflammopharmacology 2011, 19, 307–316. [Google Scholar] [CrossRef] [PubMed]
  52. Spivak, A.Y.; Keiser, J.; Vargas, M.; Gubaidullin, R.R.; Nedopekina, D.A.; Shakurova, E.R.; Khalitova, R.R.; Odinokov, V.N. Synthesis and Activity of New Triphenylphosphonium Derivatives of Betulin and Betulinic Acid against Schistosoma mansoni in vitro and in vivo. Bioorg. Med. Chem. 2014, 22, 6297–6304. [Google Scholar] [CrossRef] [PubMed]
  53. Guimarães, M.A.; de Oliveira, R.N.; Véras, L.M.C.; Lima, D.F.; Campelo, Y.D.M.; Campos, S.A.; Kuckelhaus, S.A.S.; Pinto, P.L.S.; Eaton, P.; Mafud, A.C.; et al. Anthelmintic Activity In vivo of Epiisopiloturine against Juvenile and Adult Worms of Schistosoma mansoni. PLoS Negl. Trop. Dis. 2015, 9, e0003656. [Google Scholar] [CrossRef] [PubMed]
  54. Silva, M.P.; de Oliveira, R.N.; Mengarda, A.C.; Roquini, D.B.; Allegretti, S.M.; Salvadori, M.C.; Teixeira, F.S.; de Sousa, D.P.; Pinto, P.L.S.; da Silva Filho, A.A.; et al. Antiparasitic Activity of Nerolidol in a Mouse Model of Schistosomiasis. Int. J. Antimicrob. Agents 2017, 50, 467–472. [Google Scholar] [CrossRef] [PubMed]
  55. Abd El-Aal, N.F.; Hamza, R.S.; Harb, O. Paeoniflorin Targets Apoptosis and Ameliorates Fibrosis in Murine Schistosomiasis Mansoni: A Novel Insight. Exp. Parasitol. 2017, 183, 23–32. [Google Scholar] [CrossRef]
  56. Castro, A.P.; Kawano, T.; Spelta, L.E.W.; de Castro, A.T.; Pereira, N.A.; Couto, F.F.B.; dos Santos, M.H.; Boralli, V.B.; Marques, M.J. In vivo Schistosomicidal Activity of 7-Epiclusianone and Its Quantification in the Plasma of Healthy and Schistosoma mansoni Infected Mice Using UPLC-MS/MS. Phytomedicine 2018, 38, 66–73. [Google Scholar] [CrossRef] [PubMed]
  57. Metwally, D.M.; Al-Olayan, E.M.; Alanazi, M.; Alzahrany, S.B.; Semlali, A. Antischistosomal and Anti-Inflammatory Activity of Garlic and Allicin Compared with That of Praziquantel in vivo. BMC Complement. Altern. Med. 2018, 18, 135. [Google Scholar] [CrossRef]
  58. Guimarães, M.A.; de Oliveira, R.N.; de Almeida, R.L.; Mafud, A.C.; Sarkis, A.L.V.; Ganassin, R.; da Silva, M.P.; Roquini, D.B.; Veras, L.M.; Sawada, T.C.H.; et al. Epiisopilosine alkaloid has activity against Schistosoma mansoni in mice without acute toxicity. PLoS ONE. 2018, 13, e0196667. [Google Scholar] [CrossRef]
  59. Mengarda, A.C.; Mendonça, P.S.; Morais, C.S.; Cogo, R.M.; Mazloum, S.F.; Salvadori, M.C.; Teixeira, F.S.; Morais, T.R.; Antar, G.M.; Lago, J.H.G.; et al. Antiparasitic Activity of Piplartine (Piperlongumine) in a Mouse Model of Schistosomiasis. Acta Trop. 2020, 205, 105350. [Google Scholar] [CrossRef] [PubMed]
  60. Keiser, J.; Koch, V.; Deckers, A.; Cheung, H.T.A.; Jung, N.; Bräse, S. Naturally Occurring Cardenolides Affecting Schistosoma mansoni. ACS Infect. Dis. 2020, 6, 1922–1927. [Google Scholar] [CrossRef]
  61. Matos-Rocha, T.J.; Cavalcanti, M.G.S.; Veras, D.L.; Santos, A.F.; Freitas, C.F.; Suassuna, A.S.C.L.; Melo, E.S.; Barbosa-Filho, J.M.; Alves, L.C.; Santos, F.A.B.D. In vivo effect of essential oil of Mentha x villosa and its active compound against Schistosoma mansoni (Sambon, 1907). Braz J. Biol. 2020, 80, 582–588. [Google Scholar] [CrossRef] [PubMed]
  62. Silva, L.M.; Marconato, D.G.; da Silva, M.P.N.; Raposo, N.R.B.; Facchini, G.d.F.S.; Macedo, G.C.; Teixeira, F.D.S.; Salvadori, M.C.B.d.S.; Pinto, P.D.F.; de Moraes, J.; et al. Licochalcone A-Loaded Solid Lipid Nanoparticles Improve Antischistosomal Activity in vitro and in vivo. Nanomedicine 2021, 16, 1641–1655. [Google Scholar] [CrossRef]
  63. Silva, C.B.; Mengarda, A.C.; Rodrigues, V.C.; Cajas, R.A.; Carnaúba, P.U.; Espírito-Santo, C.; Bezerra-Filho, C.S.M.; Souza, D.P.; Moraes, J. Efficacy of Caracryl Acetate in vitro and Following Oral Administration to Mice Harboring either Prepatent or Patent Shistosoma mansoni Infections. Parasitol. Res. 2021, 120, 3837–3844. [Google Scholar] [CrossRef]
  64. Carvalho, L.S.A.; Silva, L.M.; de Souza, V.C.; da Silva, M.P.N.; Capriles, P.V.S.Z.; de Faria Pinto, P.; de Moraes, J.; Da Silva Filho, A.A. Cardamonin Presents in vivo Activity against Schistosoma mansoni and Inhibits Potato Apyrase. Chem. Biodivers. 2021, 18, e2100604. [Google Scholar] [CrossRef] [PubMed]
  65. de Carvalho, L.S.A.; de Souza, V.C.; Rodrigues, V.C.; Ribeiro, A.C.; Nascimento, J.W.L.; Capriles, P.V.S.Z.; Pinto, P.F.; de Moraes, J.; da Silva Filho, A.A. Identification of Asiaticoside from Centella erecta (Apiaceae) as Potential Apyrase Inhibitor by UF-UHPLC-MS and Its In vivo Antischistosomal Activity. Pharmaceutics 2022, 14, 1071. [Google Scholar] [CrossRef] [PubMed]
  66. Bakery, H.H.; Allam, G.A.; Abuelsaad, A.S.A.; Abdel-Latif, M.; Elkenawy, A.E.; Khalil, R.G. Anti-inflammatory, Antioxidant, Anti-fibrotic and Schistosomicidal Properties of Plumbagin in Murine Schistosomiasis. Parasite Immunol. 2022, 44, e12945. [Google Scholar] [CrossRef] [PubMed]
  67. Khalil, R.G.; Ibrahim, A.M.; Bakery, H.H. Juglone: “A Novel Immunomodulatory, Antifibrotic, and Schistosomicidal Agent to Ameliorate Liver Damage in Murine schistosomiasis Mansoni”. Int. Immunopharmacol. 2022, 113, 109415. [Google Scholar] [CrossRef]
  68. Gorabi, A.M.; Razi, B.; Aslani, S.; Abbasifard, M.; Imani, D.; Sathyapalan, T.; Sahebkar, A. Effect of curcumin on proinflammatory cytokines: A meta-analysis of randomized controlled trials. Cytokine 2021, 143, 155541. [Google Scholar] [CrossRef]
  69. Schramm, G.; Haas, H. Th2 Immune Response against Schistosoma mansoni Infection. Microbes Infect. 2010, 12, 881–888. [Google Scholar] [CrossRef]
  70. Luna, E.C.; Luna, I.S.; Scotti, L.; Monteiro, A.F.M.; Scotti, M.T.; de Moura, R.O.; Mendonca, F.J.B. Active essential oils and their components in use against neglected diseases and Arboviruses. Oxidative Med. Cell. Longev. 2019, 2019, 6587150–6587152. [Google Scholar] [CrossRef] [PubMed]
  71. Mendes, N.M.; Araújo, N.; de Souza, C.P.; Pereira, J.P.; Katz, N. Molluscacide and cercariacide activity of different species of Eucalyptus. Rev. Soc. Bras. Med. Trop. 1990, 23, 197–199. [Google Scholar] [CrossRef]
  72. de Carvalho Augusto, R.; Merad, N.; Rognon, A.; Gourbal, B.; Bertrand, C.; Djabou, N.; Duval, D. Molluscicidal and parasiticidal activities of Eryngium triquetrum essential oil on Schistosoma mansoni and its intermediate snail host Biomphalaria Glabrata, a double impact. Parasit. Vectors 2020, 13, 486. [Google Scholar] [CrossRef] [PubMed]
  73. Ribeiro, E.C.G.; Leite, J.A.C.; Luz, T.R.S.A.; Silveira, D.P.B.; Bezerra, S.A.; Frazão, G.C.C.G.; Pereira, L.P.L.A.; Guimarães Dos Santos, E.G.; Ribeiro Filho, P.R.C.F.; Soares, A.M.S.; et al. Molluscicidal activity of monoterpenes and their effects on inhibition of acetylcholinesterase activity on Biomphalaria glabrata, an intermediate host of Schistosoma mansoni. Acta Trop. 2021, 223, 106089. [Google Scholar] [CrossRef] [PubMed]
  74. Araújo, F.P.; Albuquerque, R.D.D.G.; Rangel, L.D.S.; Caldas, G.R.; Tietbohl, L.A.C.; Santos, M.G.; Ricci-Júnior, E.; Thiengo, S.; Fernandez, M.A.; Santos, J.A.A.D.; et al. Nanoemulsion containing essential oil from Xylopia ochrantha Mart. produces molluscicidal effects against different species of Biomphalaria (Schistosoma hosts). Mem. Inst. Oswaldo Cruz 2019, 114, e180489. [Google Scholar] [CrossRef]
  75. Santos-Filho, D.; Sarti, S.J.; Katz, N.; Araújo, N.; Rocha Filho, P.A.; Abreu, J.E.; Bortolin, M.E. Chemoprophylactic activity of soaps containing essential oil from the fruit of Pterodon pubescens in schistosomiasis mansoni. Mem. Inst. Oswaldo Cruz 1987, 82 (Suppl. S4), 343–345. [Google Scholar]
  76. Saleh, M.M.; Zwaving, J.H.; Malingré, T.M.; Bos, R. The essential oil of Apium graveolens var. secalinum and its cercaricidal activity. Pharm. Weekbl. Sci. 1985, 7, 277–279. [Google Scholar] [CrossRef]
  77. Naples, J.M.; Shiff, C.J.; Rosler, K.H. Schistosoma mansoni: Cercaricidal effects of Cedarwood oil and various of its components. J. Trop. Med. Hyg. 1992, 95, 390–396. [Google Scholar] [PubMed]
  78. Parreira, N.A.; Magalhães, L.G.; Morais, D.R.; Caixeta, S.C.; de Sousa, J.P.; Bastos, J.K.; Cunha, W.R.; Silva, M.L.; Nanayakkara, N.P.; Rodrigues, V.; et al. Antiprotozoal, schistosomicidal, and antimicrobial activities of the essential oil from the leaves of Baccharis dracunculifolia. Chem. Biodivers. 2010, 7, 993–1001. [Google Scholar] [CrossRef]
  79. Caixeta, S.C.; Magalhães, L.G.; de Melo, N.I.; Wakabayashi, K.A.; Aguiar Gde, P.; Mantovani, A.L.; Alves, J.M.; Oliveira, P.F.; Tavares, D.C.; Groppo, M.; et al. Chemical composition and in vitro schistosomicidal activity of the essential oil of Plectranthus neochilus grown in Southeast Brazil. Chem. Biodivers. 2011, 8, 2149–2157. [Google Scholar] [CrossRef] [PubMed]
  80. de Melo, N.I.; Magalhaes, L.G.; de Carvalho, C.E.; Wakabayashi, K.A.; de P Aguiar, G.; Ramos, R.C.; Mantovani, A.L.; Turatti, I.C.; Rodrigues, V.; Groppo, M.; et al. Schistosomicidal activity of the essential oil of Ageratum conyzoides L. (Asteraceae) against adult Schistosoma mansoni worms. Molecules 2011, 16, 762–773. [Google Scholar] [CrossRef] [PubMed]
  81. de Melo, N.I.; Mantovani, A.L.; de Oliveira, P.F.; Groppo, M.; Filho, A.A.; Rodrigues, V.; Cunha, W.R.; Tavares, D.C.; Magalhães, L.G.; Crottii, A.E. Antischistosomal and Cytotoxic Effects of the Essential Oil of Tetradenia riparia (Lamiaceae). Nat. Prod. Commun. 2015, 10, 1627–1630. [Google Scholar] [CrossRef] [PubMed]
  82. Matos-Rocha, T.J.; dos Santos Cavalcanti, M.G.; Barbosa-Filho, J.M.; Lúcio, A.S.; Veras, D.L.; Feitosa, A.P.; de Siqueira Júnior, J.P.; de Almeida, R.N.; Marques, M.O.; Alves, L.C.; et al. In vitro evaluation of schistosomicidal activity of essential oil of Mentha x villosa and some of its chemical constituents in adult worms of Schistosoma mansoni. Planta Med. 2013, 79, 1307–1312. [Google Scholar] [CrossRef]
  83. de Oliveira, R.N.; Rehder, V.L.; Santos Oliveira, A.S.; Júnior, Í.M.; de Carvalho, J.E.; de Ruiz, A.L.; Jeraldo, V.d.L.; Linhares, A.X.; Allegretti, S.M. Schistosoma mansoni: In vitro schistosomicidal activity of essential oil of Baccharis trimera (less) DC. Exp. Parasitol. 2012, 132, 135–143. [Google Scholar] [CrossRef] [PubMed]
  84. Matos-Rocha, T.J.; Cavalcanti, M.G.; Veras, D.L.; Feitosa, A.P.; Gonçalves, G.G.; Portela-Junior, N.C.; Lúcio, A.S.; Silva, A.L.; Padilha, R.J.; Marques, M.O.; et al. Ultrastructural changes in Schistosoma mansoni male worms after in vitro incubation with the essential oil of Mentha x villosa Huds. Rev. Inst. Med. Trop. Sao Paulo 2016, 58, 1–6. [Google Scholar] [CrossRef] [PubMed]
  85. Wakabayashi, K.A.; de Melo, N.I.; Aguiar, D.P.; de Oliveira, P.F.; Groppo, M.; da Silva Filho, A.A.; Rodrigues, V.; Cunha, W.R.; Tavares, D.C.; Magalhães, L.G.; et al. Anthelmintic effects of the essential oil of fennel (Foeniculum vulgare Mill., Apiaceae) against Schistosoma mansoni. Chem. Biodivers. 2015, 12, 1105–1114. [Google Scholar] [CrossRef] [PubMed]
  86. Martins, M.H.; Fracarolli, L.; Vieira, T.M.; Dias, H.J.; Cruz, M.G.; Deus, C.C.; Nicolella, H.D.; Stefani, R.; Rodrigues, V.; Tavares, D.C.; et al. Schistosomicidal Effects of the Essential Oils of Citrus limonia and Citrus reticulata against Schistosoma mansoni. Chem. Biodivers. 2017, 14, e1600194. [Google Scholar] [CrossRef] [PubMed]
  87. Soares, M.H.; Dias, H.J.; Vieira, T.M.; de Souza, M.G.M.; Cruz, A.F.F.; Badoco, F.R.; Nicolella, H.D.; Cunha, W.R.; Groppo, M.; Martins, C.H.G.; et al. Chemical Composition, Antibacterial, Schistosomicidal, and Cytotoxic Activities of the Essential Oil of Dysphania ambrosioides (L.) Mosyakin & Clemants (Chenopodiaceae). Chem. Biodivers. 2017, 14, e1700149. [Google Scholar] [CrossRef]
  88. Rizk, M.; Ibrahim, N.; El-Rigal, N. Comparative in vivo antioxidant levels in Schistosoma mansoni infected mice treated with praziquantel or the essential oil of Melaleuca armillaris leaves. Pak. J. Biol. Sci. 2012, 15, 971–978. [Google Scholar] [CrossRef]
  89. Mafud, A.C.; Ferreira, L.G.; Mascarenhas, Y.P.; Andricopulo, A.D.; de Moraes, J. Discovery of novel antischistosomal agents by molecular modeling approaches. Trends Parasitol. 2016, 32, 874–886. [Google Scholar] [CrossRef]
  90. Moreira-Filho, J.T.; Silva, A.C.; Dantas, R.F.; Gomes, B.F.; Souza Neto, L.R.; Brandao-Neto, J.; Owens, R.J.; Furnham, N.; Neves, B.J.; Silva-Junior, F.P.; et al. Schistosomiasis drug discovery in the era of automation and artificial intelligence. Front. Immunol. 2021, 12, 642383. [Google Scholar] [CrossRef]
  91. Yu, W.; Weber, D.J.; MackKerell, A.D., Jr. Computer-Aided Drug Design: An update. Methods Mol. Biol. 2023, 2601, 123–152. [Google Scholar] [CrossRef] [PubMed]
  92. Kwon, S.; Bae, H.; Jo, J.; Yoon, S. Comprehensive ensemble in QSAR prediction for drug discovery. BMC Bioinform. 2019, 20, 521. [Google Scholar] [CrossRef]
  93. Berriman, M.; Haas, B.J.; LoVerde, P.T.; Wilson, R.A.; Dillon, G.P.; Cerqueira, G.C.; Mashiyama, S.T.; Al-Lazikani, B.; Andrade, L.F.; Ashton, P.D.; et al. The genome of the blood fluke Schistosoma mansoni. Nature 2009, 460, 352–358. [Google Scholar] [CrossRef]
  94. Buddenborg, S.K.; Tracey, A.; Berger, D.J.; Lu, Z.; Doyle, S.R.; Fu, B.; Yang, F.; Reid, A.J.; Rodgers, F.H.; Rinaldi, G.; et al. Assembled chromosomes of the blood fluke Schistosoma mansoni provide insight into the evolution of its ZW sex-determination system. BioRxiv 2021, 13, 456314. [Google Scholar] [CrossRef]
  95. Lund, A.J.; Wade, K.J.; Nikolakis, Z.L.; Ivey, K.N.; Perry, B.W.; Pike, H.N.C.; Paull, S.H.; Liu, Y.; Castoe, T.A.; Pollock, D.D.; et al. Integrating genomic and epidemiologic data to accelerate progress toward schistosomiasis elimination. Elife 2022, 11, e79320. [Google Scholar] [CrossRef] [PubMed]
  96. Wangwiwatsin, A.; Protasio, A.V.; Wilson, S.; Owusu, C.; Holroyd, N.E.; Sanders, M.J.; Keane, J.; Doenhoff, M.J.; Rinaldi, G.; Berriman, M. Transcriptome of the parasitic flatworm Schistosoma mansoni during intra-mammalian development. PLoS Negl. Trop. Dis. 2020, 14, e0007743. [Google Scholar] [CrossRef]
  97. Cheuka, P.M. Drug discovery and target identification against schistosomiasis: A reality check on progress and future prospects. Curr. Top. Med. Chem. 2022, 22, 1595–1610. [Google Scholar] [CrossRef]
  98. Ali, Z.; Hayat, M.F.; Shaukat, K.; Alam, T.M.; Hameed, I.A.; Luo, S.; Basheer, S.; Ayadi, M.; Ksibi, A. A proposed framework for early prediction of schistosomiasis. Diagnostics 2022, 12, 3138. [Google Scholar] [CrossRef] [PubMed]
  99. Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; et al. Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596, 583–589. [Google Scholar] [CrossRef]
  100. Akachukwu, I.; Olubiyi, O.O.; Kosisochukwu, A.; John, M.C.; Justina, N.N. Structure-based study of natural products with anti-schistosoma activity. Curr. Comput. Aided Drug Des. 2017, 13, 91–100. [Google Scholar] [CrossRef]
  101. de Menezes, R.P.B.; Viana, D.O.; Muratov, E.; Scotti, L.; Scotti, M.T. Computer-assisted discovery of alkaloids with schistosomicidal activity. Curr. Issues Mol. Biol. 2022, 44, 383–408. [Google Scholar] [CrossRef] [PubMed]
  102. Neves, B.J.; Dantas, R.F.; Senger, M.R.; Melo-Filho, C.C.; Valente, W.C.G.; de Almeida, A.C.M.; Rezende-Neto, J.M.; Lima, E.F.C.; Paveley, R.; Furnham, N.; et al. Discovery of new anti-schistosomal hits by integration of QSAR- based virtual screening and high content screening. J. Med. Chem. 2016, 59, 7075–7088. [Google Scholar] [CrossRef] [PubMed]
  103. Gallinger, T.L.; Aboagye, S.Y.; Obermann, W.; Weiss, M.; Grünweller, A.; Unverzagt, C.; Williams, D.L.; Schlitzer, M.; Haeberlein, S. First in silico screening of insect molecules for identification of novel anti-parasitic compounds. Pharmaceuticals 2022, 15, 119. [Google Scholar] [CrossRef] [PubMed]
  104. Moreira, B.P.; Weber, M.H.W.; Haeberlein, S.; Mokosch, A.S.; Spengler, B.; Grevelding, C.G.; Falcone, F.H. Drug repurposing and de novo drug discovery of protein kinase inhibitors as new drugs against schistosomiasis. Molecules 2022, 27, 1414. [Google Scholar] [CrossRef]
  105. Marcellino, C.; Gut, J.; Lim, K.C.; Singh, R.; Mckerrow, J.; Sakanari, J. WormAssay: A novel computer application for whole-plate motion-based screening of macroscopic parasites. PLoS Negl. Trop. Dis. 2012, 6, e1494. [Google Scholar] [CrossRef]
  106. Mathavan, I.; Liu, L.J.; Robinson, S.W.; El-Sakkary, N.; Elatico, A.J.J.; Gomez, D.; Nellas, R.; Owens, R.J.; Zuercher, W.; Navratilova, I.; et al. Identification of inhibitors of the Schistosoma mansoni VKR2 kinase domain. ACS Med. Chem. Lett. 2022, 13, 1715–1722. [Google Scholar] [CrossRef] [PubMed]
  107. Simoben, C.V.; Robaa, D.; Chakrabarti, A.; Schmidtkunz, K.; Marek, M.; Lancelot, J.; Kannan, S.; Melesina, J.; Shaik, T.B.; Pierce, R.J.; et al. A novel class of Schistosoma mansoni histone deacetylase 8 (HDAC8) inhibitors identified by structure-based virtual screening and in vitro testing. Molecules 2018, 23, 566. [Google Scholar] [CrossRef]
  108. Berger, D.J.; Crellen, T.; Lamberton, P.H.L.; Allan, F.; Tracey, A.; Noonan, J.D.; Kabatereine, N.B.; Tukahebwa, E.M.; Adriko, M.; Holroyd, N.; et al. Whole-Genome Sequencing of Schistosoma mansoni Reveals Extensive Diversity with Limited Selection despite Mass Drug Administration. Nat. Commun. 2021, 12, 4776. [Google Scholar] [CrossRef] [PubMed]
  109. Vianney, T.J.; Berger, D.J.; Doyle, S.R.; Sankaranarayanan, G.; Serubanja, J.; Nakawungu, P.K.; Besigye, F.; Sanya, R.E.; Holroyd, N.; Allan, F.; et al. Genome-Wide Analysis of Schistosoma mansoni Reveals Limited Population Structure and Possible Praziquantel Drug Selection Pressure within Ugandan Hot-Spot Communities. PLoS Negl. Trop. Dis. 2022, 16, e0010188. [Google Scholar] [CrossRef] [PubMed]
  110. Kenney, E.T.; Mann, V.H.; Ittiprasert, W.; Rosa, B.A.; Mitreva, M.; Bracken, B.K.; Loukas, A.; Brindley, P.J.; Sotillo, J. Differential Excretory/Secretory Proteome of the Adult Female and Male Stages of the Human Blood Fluke, Schistosoma mansoni. Front. Parasitol. 2022, 1, 950744. [Google Scholar] [CrossRef]
  111. Abou-El-Naga, I.F.; Amer, E.I.; Boulos, L.M.; El-Faham, M.H.; Abou Seada, N.M.; Younis, S.S. Biological and Proteomic Studies of Schistosoma mansoni with Decreased Sensitivity to Praziquantel. Comp. Immunol. Microbiol. Infect. Dis. 2019, 66, 101341. [Google Scholar] [CrossRef] [PubMed]
  112. Pinto-Almeida, A.; Mendes, T.M.F.; Ferreira, P.; Abecasis, A.B.; Belo, S.; Anibal, F.F.; Allegretti, S.M.; Galinaro, C.A.; Carrilho, E.; Afonso, A. A Comparative Proteomic Analysis of Praziquantel-Susceptible and Praziquantel-Resistant Schistosoma mansoni Reveals Distinct Response between Male and Female Animals. Front. Trop. Dis. 2021, 2, 664642. [Google Scholar] [CrossRef]
  113. Protasio, A.V.; Tsai, I.J.; Babbage, A.; Nichol, S.; Hunt, M.; Aslett, M.A.; De Silva, N.; Velarde, G.S.; Anderson, T.J.C.; Clark, R.C.; et al. A Systematically Improved High Quality Genome and Transcriptome of the Human Blood Fluke Schistosoma mansoni. PLoS Negl. Trop. Dis. 2012, 6, e1455. [Google Scholar] [CrossRef] [PubMed]
  114. Sanchez, M.C.; Cupit, P.M.; Bu, L.; Cunningham, C. Transcriptomic Analysis of Reduced Sensitivity to Praziquantel in Schistosoma mansoni. Mol. Biochem. Parasitol. 2019, 228, 6–15. [Google Scholar] [CrossRef]
  115. Wendt, G.R.; Reese, M.L.; Collins, J.J., 3rd. SchistoCyte Atlas: A Single-Cell Transcriptome Resource for Adult Schistosomes. Trends Parasitol. 2021, 37, 585–587. [Google Scholar] [CrossRef]
  116. Soria, C.L.D.; Attenborough, T.; Lu, Z.; Graham, J.; Hall, C.; Thompson, S.; Andrews, T.G.R.; Rawlinson, K.A.; Berriman, M.; Rinaldi, G. Single Cell Transcriptomics of the Human Parasite Schistosoma mansoni First Intra-Molluscan Stage Reveals Tentative Tegumental and Stem Cell Regulators. bioRxiv 2023. [Google Scholar] [CrossRef]
  117. Padalino, G.; Ferla, S.; Brancale, A.; Chalmers, I.W.; Hoffmann, K.F. Combining Bioinformatics, Cheminformatics, Functional Genomics and Whole Organism Approaches for Identifying Epigenetic Drug Targets in Schistosoma mansoni. Int. J. Parasitol. Drugs Drug Resist. 2018, 8, 559–570. [Google Scholar] [CrossRef]
Figure 1. Chemical structures of natural products with antischistosomal activity demonstrated either in in vitro assays and/or murine models of infection.
Figure 1. Chemical structures of natural products with antischistosomal activity demonstrated either in in vitro assays and/or murine models of infection.
Molecules 28 06807 g001
Figure 2. Main mechanisms of action of natural products against adult worms of S. mansoni. In general, natural products induced tegument damage in schistosomes associated with body deformation, morphological disfiguring of the oral and ventral suckers, extensive sloughing, loss of tubercles, and shrinking (1). In addition, some natural products, such as licochalcone A, promoted swelling and degeneration of mitochondria and nuclear chromatin condensation, which correlated with increased superoxide anion levels and decreased superoxide dismutase activity (2). Some natural products, mainly chalcones, inhibited S. mansoni ATPase and ADPase activity (3). The reactive oxygen species (ROS) are mainly produced at the electron transport chain (ETC) in the mitochondria, which are formed by transmembrane protein complexes (I–IV). During transportation, leaked electrons interact with oxygen to form superoxide anions (O2-) at complexes I and III. These complexes are the major source of superoxide and hydrogen peroxide (H2O2) since the O2- released can be reduced into H2O2 through a reaction catalyzed by superoxide dismutase (SOD) (Tirichen et al., 2021 [40]; Brand, 2016 [41]). The overproduction of superoxide anions leads to oxidative stress and activates transcription factors such as NF-κB and AP-1. Moreover, increased ROS in mitochondria can induce the release of transmembrane proteins such as cytochrome c, an electron carrier between complexes III and IV, into the cytosol that triggers the apoptotic machinery of the cell (Tirichen et al., 2021 [40]; Guerra-Castellano, 2018 [42]).
Figure 2. Main mechanisms of action of natural products against adult worms of S. mansoni. In general, natural products induced tegument damage in schistosomes associated with body deformation, morphological disfiguring of the oral and ventral suckers, extensive sloughing, loss of tubercles, and shrinking (1). In addition, some natural products, such as licochalcone A, promoted swelling and degeneration of mitochondria and nuclear chromatin condensation, which correlated with increased superoxide anion levels and decreased superoxide dismutase activity (2). Some natural products, mainly chalcones, inhibited S. mansoni ATPase and ADPase activity (3). The reactive oxygen species (ROS) are mainly produced at the electron transport chain (ETC) in the mitochondria, which are formed by transmembrane protein complexes (I–IV). During transportation, leaked electrons interact with oxygen to form superoxide anions (O2-) at complexes I and III. These complexes are the major source of superoxide and hydrogen peroxide (H2O2) since the O2- released can be reduced into H2O2 through a reaction catalyzed by superoxide dismutase (SOD) (Tirichen et al., 2021 [40]; Brand, 2016 [41]). The overproduction of superoxide anions leads to oxidative stress and activates transcription factors such as NF-κB and AP-1. Moreover, increased ROS in mitochondria can induce the release of transmembrane proteins such as cytochrome c, an electron carrier between complexes III and IV, into the cytosol that triggers the apoptotic machinery of the cell (Tirichen et al., 2021 [40]; Guerra-Castellano, 2018 [42]).
Molecules 28 06807 g002
Figure 3. Main outcomes of in vivo treatments with natural products. Following intraperitoneal or oral administration, natural products have demonstrated an ability to reduce Schistosoma egg deposition in tissue and worm burden, leading to a reduction in the size of hepatic granulomas and fibrotic areas. Some compounds, such as nerolidol and piplartine, were observed to induce tegument damage as a mechanism of worm elimination. Moreover, plumbagin, curcumin, and other natural products exhibited different immunomodulatory properties by modulating cytokine production associated with Th1, Th2, and Th17 profiles.
Figure 3. Main outcomes of in vivo treatments with natural products. Following intraperitoneal or oral administration, natural products have demonstrated an ability to reduce Schistosoma egg deposition in tissue and worm burden, leading to a reduction in the size of hepatic granulomas and fibrotic areas. Some compounds, such as nerolidol and piplartine, were observed to induce tegument damage as a mechanism of worm elimination. Moreover, plumbagin, curcumin, and other natural products exhibited different immunomodulatory properties by modulating cytokine production associated with Th1, Th2, and Th17 profiles.
Molecules 28 06807 g003
Table 1. In vitro antischistosomal activity of compounds isolated from natural sources.
Table 1. In vitro antischistosomal activity of compounds isolated from natural sources.
MoleculesConcentrationsMain ResultsReferences
Dermaseptin 0125, 50, 75, 100, 150,
and 200 µg/mL
Dermaseptin 01 reduced motility and induced death in adult worms of S. mansoni at concentrations between 50 and 200 µg/mL. In addition, Dermaseptin 01 reduced the egg output of paired female worms and induced morphological alterations in the tegument of S. mansoni[29]
Betulin, Oleanolic acid, Ursolic acid, Quercetin 3-O-β-d-rhamnoside, Quercetin 3-O-β-d-glucoside, Quercetin 3-O-β-d-glucopyranosyl-(1-2)- α-l-rhamnopyranoside, and Isorhamnetin 3-O-β-d-glucopyranosyl-(1-2)-α-l-rhamnopyranoside50, 100, and 200 µMNatural products reduced motor activity and caused death in adult S. mansoni worms[22]
Pirplatine7.5, 15, 30, and 60 µMPiplartine treatment resulted in the death of all schistosomula in a concentration- and time-dependent manner. Microscopic observation revealed extensive tegumental destruction, including blebbing, granularity, and shortened S. mansoni schistosomula body length.[23]
Balsaminol F and Karavilagenin C10, 25, 50, and 100 µMBalsaminol F and Karavilagenin presented LC50 values of 14.7 and 28.9 µM, respectively, against 56-day-old adult S. mansoni. In addition, at 10–50 µM, both compounds caused significantly reduced worm motor activity and significantly decreased egg production. At 10–100 µM, both triterpenes separated adult worm pairs into males and females after 24 h[27]
(+)-limonene epoxide12.5, 25, 50, and 75 µg/mLTreatment with compound reduced motility and induced death in adult S. mansoni worms at concentrations ≥25 µg/mL. Microscopic analysis revealed (+)-limonene epoxide mediated worm killing in association with tegumental destruction[30]
Hesperidin50, 100, and 200 µg/mLHesperidin, at 200 µg/mL, caused 100% mortality in 56-day-old adult worms within 72 h, with partial tegumental alterations observed in 10% of worms[43]
N-[7-(30,40-methylenedioxyphenyl)-2(Z),4(Z)-heptadienoyl] pyrrolidine10, 25, 50, and 100 µMThe isolated compound N-[7-(3′,4′-methylenedioxyphenyl)-2(Z),4(Z)-heptadienoyl] pyrrolidine promoted death of all adult worms of S. mansoni at 100 µM after 24 h of treatment[31]
Phytol12.5, 25, 50, 75, and 100 µg/mLTreatment with phytol reduced worm motor activity and caused death. Confocal laser scanning microscopy analysis revealed extensive tegumental alterations in a concentration-dependent manner (50 to 100 µg/mL). Additionally, sublethal doses of phytol (25 µg/mL) reduced numbers of Schistosoma mansoni eggs[44]
Diethyl 4-phenyl-2,6-dimethyl-3,5-pyridinedicarboxylate1, 10, and 100 µg/mLThe alkaloid promoted the inhibition of movement and death in S. mansoni adult worms, accompanied by the formation of vesicles and vacuolization. In addition, the alkaloid exhibited a potent cercaricidal activity (LC100 = 2 μg/mL) as well as activity against adult snails (LC90 = 36.43 μg/ mL)[32]
Nerolidol15.6, 31.2, 62.5, 125, and 250 µMNerolidol reduced motor activity and caused death in adult S. mansoni worms. In addition, morphological alterations were observed in the tegument of worms (disintegration, sloughing, and surface erosion)[26]
Licoflavone B5, 10, 25, 50, and 100 µMLicoflavone B (25 to 100 µM) caused 100% mortality, tegumental alterations, and reduced oviposition and motor activity in all adult worms, without affecting mammalian Vero cells. Licoflavone B also highly inhibited S. mansoni ATPase (IC50 of 23.78 µM) and ADPase (IC50 of 31.50 µM) activity[33]
Streptomycete-derived compound SF2446A20.5–10 µMTreatment with 100 µM of SF2446A2 affected the gonads by impairing oogenesis and spermatogenesis. In addition, SF2446A2 caused disruptive effects on the tegument surface of S. mansoni[45]
Cardol triene, Cardol diene, Anacardic acid triene, Cardol monoene, Anacardic acid diene, 2-methylcardol triene, and 2-methylcardol diene12.5, 25, 50, 100, and 200 µMCompounds Cardol diene and 2-methylcardol diene showed activity against S. mansoni adult worms, with LC50 values of 32.2 and 14.5 μM and selectivity indices of 6.1 and 21.2, respectively. Transmission electron microscopy revealed alterations in the tegument and mitochondrial membrane.[34]
Phytol25, 50, 75, 100,
125, and 150 µg/mL
Phytol reduced motility and induced death in adult S. mansoni worms at 150 μg/mL, with male worms more susceptible to treatment. On an ultrastructural level, phytol induced tegumental peeling, disintegration of tubercles and spines, as well as morphological disfiguring of oral and ventral suckers[35]
Series of 38 terpenes10, 20, 40, 80, 100,
and 160 µM
Only dihydrocitronellol at 100 µM presented schistosomicidal activity after the maximal screening time of 120 h. Confocal laser scanning microscopy revealed severe tegumental damage induced by dihydrocitronellol in adult schistosomes[46]
Barbatic acid0.25, 0.5, 1, 10,
25, and 100 µg/mL
Barbatic acid exhibited molluscicidal activity against snails, especially at 25 µg/mL, with 100% lethality. In addition, barbatic acid presented cercaricidal activity, completely eliminating cercariae at concentrations between 1 and 100 µg/mL[47]
Terrein, Butyrolactone I, and butyrolactone V25–1297.3 µMAll compounds reduced motility and induced death in adult S. mansoni worms at concentrations between 235.6 and 454.1 µM[28]
Licochalcone A3.125, 6,25, 12,5, 25,
50, 100, and 200 µM
Licochalcone A reduced the number of S. mansoni eggs and affected egg development in adult worms. Drastic changes in the tegument of S. mansoni adult worms and alterations in mitochondria and chromatin condensation were related to increased superoxide anion levels and decreased superoxide dismutase activity in adult S. mansoni worms[48]
A series of 15 chalcones10, 50, and 100 µMChalcones, especially 1 and 3, induced adult worm death, reduced motility, and caused changes in the tegument of adult S. mansoni worms[36]
(-) Hinoquinin, (-)-Cubebin, Yatein, 5-Methoxyyatein, Dihydrocubebin, and Dihydroclusin.10, 25, 50, and 100 µM(-) Hinoquinin, (-)-Cubebin, Yatein, and 5-Methoxyyatein decreased motor activity in adult S. mansoni worms. All compounds, except Dihydrocubebin, were found to separate adult worm pairs and reduce egg numbers after 24 h of treatment[37]
Curcumin1.56, 3.125, 6.25, 12.5,
25, 50, and 100 µg/mL
Curcumin presented LC50 values <10 µg/mL against cercariae. Treatment with curcumin affected egg-laying capacity and egg hatchability, causing death in newborns, embryos, and adult B. globrata snails.[24]
6-[8(Z)-pentadecenyl] anacardic, 6-[10(Z)-heptadecenyl] anacardic acid, and 3-[7(Z)-pentadecenyl] phenol1, 10, and 100 µMAll compounds presented activity against S. mansoni, killing 100% of adult S. mansoni worms at 100 µM[38]
Anemonin1 and 10 µMAnemonin demonstrated activity against adult S. mansoni and newly transformed schistosomules (49% activity against adult S. mansoni at 10 µM and 41% activity against newly transformed schistosomules at 1 µM)[39]
ATPase, adenosine triphosphatases; LC50, lethal concentration of 50%; LC90, lethal concentration of 90%; LC100, lethal concentration of 100%.
Table 2. In vivo antischistosomal activity of compounds isolated from natural sources.
Table 2. In vivo antischistosomal activity of compounds isolated from natural sources.
MoleculesRouteDoseMain ResultsReferences
CurcuminIntraperitoneal400 mg/kg/dayCurcumin reduced worm and tissue egg burden, hepatic granuloma volume, and liver collagen content by 44.4%, 30.9%, 79%, and 38.6%, respectively[50]
CurcuminOral300 mg/kg/dayCurcumin treatment exerted antifibrotic effects in S. mansoni-infected mice[51]
PhytolOral40 mg/kg/dayA single dose of phytol (40 mg/kg) resulted in total and female worm burden reductions of 51.2% and 70.3%, respectively. Also, reduced numbers of eggs were found in feces (76.6%), with a lower frequency of immature eggs[44]
HesperidinIntraperitoneal100 mg/kg/dayReductions of 50, 45.2, 50, and 47.5% in males, females, worm pairs, and total worm burden, respectively. In addition, respective reductions, based on the number of eggs/g of tissue, of 41.5, 63.7, and 58.6% were observed in the liver, intestine, and liver/intestinal tissue combined[43]
TriphenylphosphoniumOral400 mg/kg/dayTriphenylphosphonium salts 10 and 11 resulted in low worm burden reductions against S. mansoni of 21.9% and 22.2%, respectively. Both compounds were well-tolerated by mice[52]
EpiisopiloturineOral40, 100, and 300 mg/Kg/dayTreatment with epiisopiloturine at 40 mg/kg reduced total worm burden by 50.2%, as well as hepatosplenomegaly, egg burden in feces, and granuloma diameter. Electron microscopy revealed a loss of important features in the parasite tegument[53]
NerolidolOral100, 200, and 400 mg/kg/dayNerolidol (100, 200, or 400 mg/kg) reduced worm burden and egg production in mice infected with adult schistosomes. Treatment with the highest concentration reduced total worms by 70.06% and immature eggs by 84.6%. Microscopic observations revealed that nerolidol-mediated worm killing was associated with tegumental damage[54]
PaeoniflorinOral50 mg/kg/dayPaeoniflorin treatment decreased worm burden, as well as immature and mature eggs, with reductions in hepatic granuloma size and fibrotic areas[55]
7-epiclusianoneOral100 or 300 mg/kg/day7-epiclusianone showed significant schistosomicidal in vivo activity following treatment with 300 mg/kg for 5 days[56]
AllicinOral0.5 μM/mouseProphylactic administration of allicin in infected mice significantly reduced worm burden. Serum concentrations of liver fibrosis markers and proinflammatory cytokines were also reduced[57]
Series of 15 chalconesOral400 mg/Kg/dayChalcones 1 and 3 demonstrated moderate schistosomicidal activity with total worm burden significantly reduced by 32.8% and 31.8%, respectively, at a single oral dose (400 mg/kg)[36]
Epiisopilosine alkaloidOral100 or 400 mg/Kg/dayA single dose of epiisopilosine significantly decreased total worm load by 57.78 and 60.61% at doses of 400 and 100 mg/Kg, respectively. In addition, epiisopilosine significantly reduced eggs number and decreased hepatosplenomegaly[58]
PiplartineOral100, 200 or 400 mg/kg/dayTreatment with the highest piplartine dose (400 mg/kg) caused a significant (60.4%) reduction in total worm burden in mice harboring adult parasites. Microscopy revealed substantial tegumental alterations in parasites recovered from mice[59]
Gomphoside monoacetate and UscharinOral10 mg/kg/dayOnly gomphoside monoacetate (10 mg/kg) demonstrated activity against S. mansoni, with a low worm burden reduction of 38%[60]
RotundifoloneOral35.9, 70.9 and
141.9 mg/Kg/day
Rotundifolone (141.9 mg/kg) significantly reduced fluke burden by 74.48%. Marked reductions in liver, intestinal, and fecal fluke burden, together with changes in the oogram pattern were observed. Treatment affected the viability of both mature and immature eggs[61]
Licochalcone AOral; intraperitoneal1.5 or 2.5 mg/kg/day (oral); 25 mg/kg/day (intraperitoneal)Oral treatment with L-SLNs decreased worm burden. However, under intraperitoneal administration, both free licochalcone A and L-SLNs significantly decreased worm burden and intestinal egg load[62]
Carvacryl acetateOral100, 200, or 400 mg/kg/dayCarvacryl acetate (400 mg/kg) showed moderate efficacy against S. mansoni, with slightly reduced worm burden (32–40%). Egg production was markedly reduced (70–80%)[63]
CardamoninOral400 mg/kg/dayOral treatment with cardamonin (400 mg/kg) demonstrated efficacy against S. mansoni, with decreased total worm load in 46.8% of mice and a 54.5% reduction in egg numbers[64]
AsiaticosideOral400 mg/kg/dayA single oral dose (400 mg/kg) of asiaticoside presented significant in vivo antischistosomal efficacy, markedly decreasing total worm and egg burden[65]
PlumbaginIntraperitoneal20 mg/kg/dayMice treated with plumbagin (20 mg/kg) showed reductions of 64.28% and 59.88% in male and female worms, respectively. Plumbagin treatment also alleviated schistosome-induced hepatosplenomegaly and reduced hepatic granuloma and liver collagen content[66]
JugloneIntraperitoneal2 mg/Kg/dayTreatment with the compound reduced male and female worms by 63.1% and 52.1%, respectively. The number of eggs/g of tissue in the liver and intestine were also reduced. Juglone decreased hepatic granuloma size and collagen fiber deposition. Mice treated with juglone presented significantly lower levels of IL-4, IL-13, IL-37, TNF-α, TGF-β, and IFN-γ than PZQ mice[67]
L-SLNs, LicoA-loaded solid lipid nanoparticles.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Azevedo, C.M.; Meira, C.S.; da Silva, J.W.; Moura, D.M.N.; de Oliveira, S.A.; da Costa, C.J.; Santos, E.d.S.; Soares, M.B.P. Therapeutic Potential of Natural Products in the Treatment of Schistosomiasis. Molecules 2023, 28, 6807. https://doi.org/10.3390/molecules28196807

AMA Style

Azevedo CM, Meira CS, da Silva JW, Moura DMN, de Oliveira SA, da Costa CJ, Santos EdS, Soares MBP. Therapeutic Potential of Natural Products in the Treatment of Schistosomiasis. Molecules. 2023; 28(19):6807. https://doi.org/10.3390/molecules28196807

Chicago/Turabian Style

Azevedo, Carine Machado, Cássio Santana Meira, Jaqueline Wang da Silva, Danielle Maria Nascimento Moura, Sheilla Andrade de Oliveira, Cícero Jádson da Costa, Emanuelle de Souza Santos, and Milena Botelho Pereira Soares. 2023. "Therapeutic Potential of Natural Products in the Treatment of Schistosomiasis" Molecules 28, no. 19: 6807. https://doi.org/10.3390/molecules28196807

Article Metrics

Back to TopTop