Review

Antiparasitic properties of curcumin: A review

  • Received: 28 September 2018 Accepted: 21 December 2018 Published: 25 December 2018
  • Medicinal plants are known as one of the most effective approaches to overcome parasitic infections, which has been used several years ago. Curcuma longa (C. longa) that is commonly used as a food additive in cooking. Curcumin as the major compound extracted from C. longa serves for the various therapeutic and preventive purposes. The present review paper is aimed to investigate the antiparasitic effects of curcumin reported in recent years. The data was collected from several databanks including ISI, Google Scholar, Pubmed, Scopus, and SID (Scientific Information Database, Iran). After a primary study of the retrieved data, the most relevant literature was subjectively classified based on the type of parasite. Then, the effect of curcumin treatment on various parasites was assessed regarding the kind of parasite. According to the results, curcumin manifested a high potential to serve as an effective drug against various parasites. Therefore, further studies in detail on curcumin might offer a new perspective that helps to design efficient formulations for hampering the infections caused by parasites in both human and animals.

    Citation: Kourosh Cheraghipour, Abdolrazagh Marzban, Behrouz Ezatpour, Sayyad Khanizadeh, Javad Koshki. Antiparasitic properties of curcumin: A review[J]. AIMS Agriculture and Food, 2018, 3(4): 561-578. doi: 10.3934/agrfood.2018.4.561

    Related Papers:

    [1] Yu Na Jung, Jungil Hong . Changes in chemical properties and bioactivities of turmeric pigments by photo-degradation. AIMS Agriculture and Food, 2021, 6(2): 754-767. doi: 10.3934/agrfood.2021045
    [2] R. Amilia Destryana, Teti Estiasih, Sukardi, Dodyk Pranowo . The potential uses of Galangal (Alpinia sp.) essential oils as the sources of biologically active compounds. AIMS Agriculture and Food, 2024, 9(4): 1064-1109. doi: 10.3934/agrfood.2024057
    [3] Andrea Ertani, Ornella Francioso, Serenella Nardi . Mini review: fruit residues as plant biostimulants for bio-based product recovery. AIMS Agriculture and Food, 2017, 2(3): 251-257. doi: 10.3934/agrfood.2017.3.251
    [4] Thi Thuy Le, Trung Kien Nguyen, Nu Minh Nguyet Ton, Thi Thu Tra Tran, Van Viet Man Le . Quality of cookies supplemented with various levels of turmeric by-product powder. AIMS Agriculture and Food, 2024, 9(1): 209-219. doi: 10.3934/agrfood.2024012
    [5] Zipora Tietel, Devanesan Arul Ananth, Thilagar Sivasudha, Liron Klipcan . Cassia auriculata L.–A mini review of phytochemical compounds and their antidiabetic mechanisms. AIMS Agriculture and Food, 2024, 9(1): 374-392. doi: 10.3934/agrfood.2024022
    [6] Joshua D. Klein, Amanda Firmansyah, Nurhaya Panga, Waffa Abu-Aklin, Miriam Dekalo-Keren, Tanya Gefen, Ronit Kohen, Yonit Raz Shalev, Nativ Dudai, Lea Mazor . Seed treatments with essential oils protect radish seedlings against drought. AIMS Agriculture and Food, 2017, 2(4): 345-353. doi: 10.3934/agrfood.2017.4.345
    [7] JoséIgnacio López-Sánchez, Diego A. Moreno, Cristina García-Viguer . D-pinitol, a highly valuable product from carob pods: Health-promoting effects and metabolic pathways of this natural super-food ingredient and its derivatives. AIMS Agriculture and Food, 2018, 3(1): 41-63. doi: 10.3934/agrfood.2018.1.41
    [8] Abdul Razak Ahmed, Samuel Obeng Apori, Abdul Aziz Karim . Mealybug vectors: A review of their transmission of plant viruses and their management strategies. AIMS Agriculture and Food, 2023, 8(3): 736-761. doi: 10.3934/agrfood.2023040
    [9] Mohsen Ahmadi Lashaki, Shahram Sedaghathoor, Sepideh Kalatehjari, Davood Hashemabadi . The physiological and growth response of Petunia hybrida, Tagetes erecta, and Calendula officinalis to plant and human steroids. AIMS Agriculture and Food, 2018, 3(2): 85-96. doi: 10.3934/agrfood.2018.2.85
    [10] Ebrahim Falahi, Zohre Delshadian, Hassan Ahmadvand, Samira Shokri Jokar . Head space volatile constituents and antioxidant properties of five traditional Iranian wild edible plants grown in west of Iran. AIMS Agriculture and Food, 2019, 4(4): 1034-1053. doi: 10.3934/agrfood.2019.4.1034
  • Medicinal plants are known as one of the most effective approaches to overcome parasitic infections, which has been used several years ago. Curcuma longa (C. longa) that is commonly used as a food additive in cooking. Curcumin as the major compound extracted from C. longa serves for the various therapeutic and preventive purposes. The present review paper is aimed to investigate the antiparasitic effects of curcumin reported in recent years. The data was collected from several databanks including ISI, Google Scholar, Pubmed, Scopus, and SID (Scientific Information Database, Iran). After a primary study of the retrieved data, the most relevant literature was subjectively classified based on the type of parasite. Then, the effect of curcumin treatment on various parasites was assessed regarding the kind of parasite. According to the results, curcumin manifested a high potential to serve as an effective drug against various parasites. Therefore, further studies in detail on curcumin might offer a new perspective that helps to design efficient formulations for hampering the infections caused by parasites in both human and animals.


    To date, numerous studies have been conducting on emerging diseases in developing countries that caused high mortality. Nevertheless, a large number of patients still die due to lack of sufficient medication and also the shortage of health-care service. Besides, emerging types of parasites facilitate recurrence of the parasitic diseases including new drug-resistant variants [1,2,3].

    Turmeric is one of the most consuming medicinal plants belonging to the family Zingiberaceae, Curcuma genus, which is commonly known as Curcuma longa (C. longa). Turmeric extract is thoroughly used in traditional medicine for treating psoriasis, teeth, gum pains, snake bite, and scorpion sting [4]. The therapeutic properties of C. longa are attributed to existing the polyphenolic curcuminoid compounds, especially diferuloylmethane or curcumin. This compound commonly presents in the rhizomes of C. longa and other Curcuma species [5,6]. This compound is a yellow-colored and crystallizable powder with high hydrophobicity that shows poor solubility in aqueous phase [7]. Curcumin possesses the remarkable potential to treat some types of skin inflammations, which in the past was used as an efficient drug for psoriasis therapy [8].

    Moreover, curcumin features some biological activity such as anti-inflammation, antioxidant, antiviral, and antimicrobial properties [9]. Due to the low solubility of curcumin in water as well as its instability in some physiological conditions, oral administration results in its poor absorption through the intestinal tract [10]. On the other hand, because of the fast metabolism of curcumin and eliminating its metabolites from the body, its therapeutic effects have significantly declined. As a result, to improve the therapeutic efficiency of the curcumin-containing compound, plenty of attempts has been made aiming to eliminate the biological barriers that might limit its pharmacokinetic properties [11].

    As the report of WHO, many of the developing countries are confronting nutritional and health problems as well as especially parasitic infections. Additionally, the prevalence of parasitic diseases is widespread in tropical regions due to the specific climatic conditions facilitating the spread of the parasites [3,12]. In this review, those most common parasites causing the human and animal diseases have been listed in Table 1.

    Table 1.  Important parasites involved in human and animal diseases.
    Parasite type Disease type Prevalence rate Death rate Reference
    leishmania cutaneous leishmaniasis and visceral leishmaniasis 12 million cases 50,000 cases [3]
    Malaria (Plasmodium) palladium, fever and shivering, periodic fever, and intermittent (recurrent) fever 216 million cases 445,000 cases [13]
    Acanthamoeba castellanii (keratitis) keratitis and granulomatous amoebic encephalitis (GAE) Not available 95% of infected patients with GAE [14,15]
    Entamoeba histolytica bloody diarrhea, amebic dysentery, amebic liver abscesses 40–50 million cases 40,000–100,000 cases [16]
    Trichomonas vaginalis vaginitis in women, urethritis in man 174 million cases Not available [17]
    Giardia lamblia giardiasis: Diarrhea, abdominal pain, and weight loss, vomiting, blood in the stool 280 million individuals annually Not available [18,19]
    Toxoplasma gondii Toxoplasmosis is flu-like illness such as muscle aches and tender lymph nodes 200,000 cases of congenital toxoplasmosis occur a year Not available [20,21]
    Continued on next page
    Parasite type Disease type Prevalence rate Death rate Reference
    Neospora caninum spontaneous abortion in infected livestock similar to Toxoplasma gondii Not available Not available [22,23]
    Cryptosporidium parvum acute, watery, and non-bloody diarrhea, respiratory cryptosporidiosis, hepatitis, and cholecystitis 403,000 people in united states in 1993 Not available [24,25]
    Eimeria tenella Hemorrhagic cecal coccidiosis in young poultry Not available Not available [26,27]
    Trypanosoma cruzi Chagas disease in South America in human, surra, and dourine in horses, and a brucellosis-like disease in cattle Not available Not available [28,29]
    Trypanosoma brucei sleeping sickness in Africa Not available Not available [30,31]
    Schistosoma schistosomiasis is known as a parasitic flatworm with clinical symptoms of diarrhea, bloody stool or blood in the urine 252 million people worldwide in 2015 4400 to 200,000 people [32,33]
    Fasciola hepatica Gastrointestinal disorders, fever, malaise, urticaria, anemia, jaundice, and respiratory infection 2.5–17 million people and 180 million at risk of infection Not available [34]

     | Show Table
    DownLoad: CSV

    Various factors, including deforestation, migration as well as promoted tourism industry led to increasing the prevalence of parasitic diseases which reveal the necessity of using effective drugs for treating the patients and carriers [35,36]. Curcuma is among the medicinal herbs used with biological properties especially antiparasitic activity [37]. On this basis, the present review is aimed to investigate the antiparasitic properties of the curcumin and its derivatives against some important human and animal parasites.

    Figure 1.  The overview of antiparasitic effects of C. longa.

    The present review was aimed to scrutiny the use of curcumin and its combination formulations some of its combinations with other drugs for killing or inhibiting the most pathogenic parasites. Some of the related literature published during the 2011–2018 was retrieved from different electronic databases including ISI, Google Scholar, Pubmed, Scopus, and SID. The scientific and general words such as curcumin, C. longa, parasite, protozoa, and helminth were searched alone and in combination in these electronic databases. All literature was first analyzed for excluding those original articles that were related to curcumin and parasites. According to the available reports, those parasites, which were treated by various kinds of curcumin formulations were selected to discuss in this review paper.

    The literature excluded from the crude research was purified regarding relatedness of the experimental and clinical outputs that are focused on the details of the effect of curcumin or extract of C. longa (as the crude curcumin) as follows in Table 2.

    Table 2.  Various formulations of curcumin affecting parasitic pathogens.
    Parasite type Curcumin formulation Therapeutic effect Reference
    Leishmania major Combination of Aloe vera extract with animal fat and Turmeric extract The formulation containing turmeric extract treated cutaneous leishmaniasis in 40% of the patients while those patients who used glucantime were about 32.7% [38]
    Leishmania major Turmeric extract Rapid death of promastigotes occurred in the presence of 2 mg/mL of turmeric extract [39]
    Leishmania major and Leishmania donovani Curcumin in Combination with netilmicin, an aminoglycoside antibiotic Antileishmanial activity of, netilmicin combined with curcumin significantly enhanced compared to when used alone [40]
    Leishmania (L.)amazonensis The liposomal formulation of turmeric cortex of Curcuma longa The MIC of two formulations (LipoRHIC and LipoRHIWC) of curcumin was obtained 5.5 and 12.5 𝜇g/mL, respectively [41]
    Leishmania major Curcumin alone and in combination with curcumin + gallium, curcumin + indium, curcumin + Diacethyle Combination therapy of curcumin with Indium (IC50 of 26 µg/mL) and Gallium (IC50 of 32 µg/mL) was more potent than curcumin alone and in combination with diacetylcurcum against Leishmania [42]
    Leishmania donovani Nanoformulation of curcumin combined with miltefosine Combination therapy of curcumin with miltefosine exhibited a synergistic effect on both promastigotes and amastigotes under in vitro conditions [43]
    Plasmodium berghei Curcumin encapsulated to PLGA Encapsulation of curcumin in PLGA led to increasing parasite suppression bout 56.8% at 5 mg/kg of nanoformulation which was higher than in free curcumin (40.5%) at 10 mg/kg [44]
    Plasmodium vivax Ethanol extracts of Curcuma caesia and Curcuma longa These extracts showed significant parasitemia inhibitions against Chloroquine-resistant P. vivax [45]
    Plasmodium berghei Curcumin-arteether combination Prevents recrudescence through immunomodulation in Plasmodium berghei in mice [46]
    Plasmodium falciparum Curcumin (In silico simulation study) The high affinity of curcumin obtained for binding with HGPRT of PfHGPRT as two virulence factors in malaria progression [47]
    Plasmodium berghei Curcumin alone CD8 + T cell and pRBC sequestration into the brain and blood-brain barrier (BBB) breakdown [48]
    Plasmodium falciparum Curcumin-loaded in FΔF nanotubes Showed of Plasmodium falciparum inhibition (IC50, 3.0 μM) as compared to free curcumin (IC50, 13 μM) [49]
    Plasmodium yoelii Curcumin bound chitosan nanoparticles Can cross the mucosal barrier intact, inhibited parasite lysate in a dose-dependent manner in a lower IC50 value than chloroquine [50]
    Plasmodium berghei Nanoformulation of curcumin inhibit P. falciparum ten-fold more than its native counterpart in vitro [51]
    Plasmodium falciparum Curcumin alone Exhibited high antimalarial activity (IC50 ~10 μM) and lowered apoptosis in bEnd.3, a endothelialpolyoma cell line [52]
    Plasmodium falciparum Curcumin analogs In silico and in vitro study showed various functional groups of curcumin and its analogs against the PfATP6 protein [53]
    Acanthamoeba castellanii Ethanol extract of Curcuma longa Found to be more effective than chlorhexidine as a common antiseptic agent [54]
    Entamoeba histolytica Curcumin- metronidazole combination 65.5% of parasite trophozoites died in the presence of curcumin- metronidazole combination [55]
    Trichomonas vaginalis Curcumin alone 100% eradication of all trichomonal cells within 24 h was reached at a concentration of 400 μg/mL curcumin [56]
    Giardia lamblia Curcumin alone Curcumin inhibited Giardia proliferation, disrupted the cytoskeletal structures of trophozoites in the dose-dependent mode [57]
    Giardia Curcumin-loaded chitosan nanoparticles The parasite was successfully eradicated from stool and intestine by curcumin-Cs nanoparticles treatment [58]
    Toxoplasma gondii Curcumin alone Curcumin inhibited the enzymatic activity of recombinant TgGlo1 and the parasitic propagation in vitro [59]
    Neospora caninum Curcumin alone Curcumin exhibited inhibitory activity with 50% growth inhibitory concentration (IC50) of 1.1 ± 0.4 [22]
    Cryptosporidium parvum Curcumin alone The anti-cryptosporidial and antioxidant activity of curcumin against C. parvum were confirmed [12]
    Eimeria tenella Dietary Curcuma longa C. longa-induced intestinal transcriptome was mostly associated with genes mediating anti-inflammatory effects [60]
    Eimeria tenella Curcumin alone At high concentrations had considerable effects on sporozoite viability and morphology [61]
    Elmira tenella Curcumin alone Curcumin showed a significantly lower percentage of sporozoite invasion than the untreated control [62]
    Trypanosoma cruzi Curcumin combined with benznidazole Combination therapy with curcumin- benznidazole showed a drastic reduction in parasitemia, mortality, cytokines (IFNγ, IL-4, and MIP-1) and myocardial inflammation [28]
    Trypanosoma cruzi Curcumin alone Curcumin inhibited parasite invasion in fibroblasts. Besides, curcumin modulated signaling pathways involved associated with inflammation, oxidative stress, and apoptosis [63]
    Trypanosoma cruzi Curcumin analogs formulations Curcuminoid as a curcumin analog showed a potent trypanocidal activity [29]
    Trypanosoma cruzi Curcumin alone Curcumin blocked calcium-dependent NFATc1 transcription, reduced COX-2, and mPGES-1 expression, and suppressed PGE2 production in ET-1 in the parasite-infected cardiomyocytes [64]
    Schistosoma mansoni Turmeric diet Turmeric reduced Schistosoma burden, granuloma size and ameliorated the pathology effects of liver infected by the parasite. [32]
    Schistosoma mansoni Curcumin alone Curcumin-induced DNA fragmentation and increased the expression of SmCASP3/7 transcripts that involved in apoptosis in Schistosoma parasitic cells [65]
    Schistosoma mansoni Curcumin alone Curcumin increased several genes involved in embryogenesis and oogenesis such as Notch and TGF-β [66]
    Fasciola gigantica Curcumin alone A significant decrease was observed in the expression of glutathione-S-transferase, and superoxide dismutase [67]
    Fasciola species Curcumin-nisin nanoparticulate Ovicidal activity of the curcumin-nisin nanoparticles was confirmed against the Fasciola species [68]

     | Show Table
    DownLoad: CSV

    Leishmaniasis refers to parasitic diseases caused by Leishmania species that transmitted by the certain types of sandflies bite. Based on the pathological features, this disease is classified into three types, cutaneous, mucocutaneous and visceral leishmaniasis. Cutaneous type is identified with lesions on the skin, the mucocutaneous type is differentiated with lesions on the skin, mouth, and nose, while the visceral type in addition to skin lesions, is diagnosed by high fever, low red blood cells, spleen swelling and liver enlargement causing immune deficiency in some cases and it may lead to death [69]. In the last decades, Sodium stibogluconate and pentamidine are the first-generation medication used for treating leishmaniasis [70].

    However, miltefosine, paromomycin, and liposomal amphotericin B, either individually or in combined form, are the selected medication used for emergency cases as well as prevention of emersion of resistance in new Leishmania strains [1]. In this regard, several formulations of curcumin were examined to treat the leishmaniasis.

    Bahrami et al. (2011) showed that among the patients treated by a herbal compound known as plant juice combination, an ingredient of which is C. longa, the healing period for the ulcers caused by cutaneous leishmaniasis (major leishmaniasis) in a 63 to 76-day interval was about 40%; while, among those patients receiving only glucantime (Meglumine antimonate), the ulcer healing period was 32.7% [38]. Hosseini et al. (2012) reported that curcumin at concentration of 2 mg/L of total turmeric extract, compared to the concentration of 0.2 mg/L, would quickly kill the promastigotes of major leishmaniasis in 88 hours, indicating the higher quantitative effect of turmeric extract in comparison to liquorice (Glycyrrhiza glabra) as well as its parasiticidal potential at lower dosages [39]. In another study by Tiwari et al. (2017), it was shown that the combination of nanocurcumin and miltefosine could be considered as a new strategy for treatment of the Leishmania-induced diseases. The compound treatment (miltefosine and nanocurcumin) in vitro, in addition to its synergistic effects on promastigotes and amastigotes, leads to the increased phagocytic activity as well as increased reactive metabolites of oxygen and nitrogen. Furthermore, it was shown that administration of the miltefosine-curcumin nanoformulation yielded a parasitic inhibition of 85% ± 1%; thus, this treatment approach, regarding its p-value of p < 0.001, was proved to have considerably higher effectiveness compared to the administration of miltefosine alone against Leishmania donovani [43]. Amaral et al. (2014) studied anti-leishmania activity of two nonpolar fractions obtained from the turmeric cortex (TC) and turmeric without cortex (TWC). The fractions were exposed to Leishmania amazonensis for determining the inhibition potency against this parasite. For this purpose, the TC and TWC extracts were dissolved in hexane, and then the resulting solution was incorporated in a liposome, the products of which were named LipoRHIC and LipoRHIWC. The best minimum inhibitory concentration (MIC) for LipoRHIC at sub-inhibitory concentration of 2.75 µg/mL was obtained equal to IC50 value of 0.4 µg/mL for 48 h treatment, indicating the treatment's effect on emersion of membrane blebs, cell shrinkage, and surface deformation, which were accompanied by mitochondrial inflammation as well as vacuolization and complete intracellular disorganization [41]. Another study conducted by Fooladvand et al. (2013) reported that indium curcumin with IC50 value of 26 µg/mL for 24 h treatment was shown to be more effective than curcumin, gallium curcumin, and indium curcumin against leishmaniasis [42].

    Malaria is among the most important parasitic diseases, which commonly appears as an acute, in some cases malignant, and sometimes long-lasting infection associated with intermittent fever and shivering, anemia, and enlarged spleen [13]. Currently, chloroquine, sulfadoxine/pyrimethamine (SP), as well as artemisinin and its derivatives are considered as the appropriate treatments for malaria; nonetheless, drug resistance and toxicity, for most of the available drugs such as chloroquine and SP have presented serious challenges to the treatment of malaria [71].

    In this regard, the researchers are increasingly attempting to find an antimalarial drug and reduce drug resistance using Curcuma extract as well as curcumin and its derivatives. Busari et al. (2017) showed that the use of curcumin-PLGA would yield higher parasiticidal property (56.8%) against P. berghei compared to pure curcumin (40.5%); also, curcumin-PLGA at lower concentrations was shown to be better than pure curcumin regarding health and anti-plasmodium activities [44]. The ethanolic extracts C. cassia and C. longa resulted in a significantly reduced parasitemia within the ranges of 5.8–75.6% and 2–29.8%, respectively, against the blood stage of P. vivax. According to the obtained results, fractions of this plant featured antimalarial potential, and the C. caesia had the lowest parasitemia (24.4%) in addition to the inhibition of invasion of malaria parasite P. vivax to the cells [45]. Studies have shown that the simultaneous use of curcumin and artemisinin has been considered as a new synergistic compound for the treatment of P. berghei, and has also exhibited its potential for improving the immune system against recurrence of P. berghei in BALB/c mice [72]. Cui et al. (2007) showed that edible administration of curcumin (100 mg/kg) during a 5-day regime reduced 80–90% of the blood parasitemia in the mice infected by P. berghei [73]. The compound treatment with AC (arteether-curcumin) applied on the ECM (experimental cerebral malaria) mice proved to be a promising approach, such that the AC was capable of reversing all of the understudy parameters including inflammatory responses, TCD8+, and obstructive red blood cells that move toward the brain vessels and break the brain-blood obstacle. In addition to postponing the mice's death for 15–20 days, curcumin resulted in the reduced parasitemia and reduced secretion of the cytokines IFN-γ and TNF-α as well as suppression of the TCD8+ in brain and spleen [48]. Among the peptide groups responsible for drug delivery, including dehydrophenylalanine (ΔPhe), phenylalanine-α, β-dehydrophenylalanine (FΔF), arginine-α, β-dehydrophenylalanine (RΔF), valine-α, β-dehydrophenylalanine (VΔF), and methionine-α, β-dehydrophenylalanine (MΔF), which were combined with various nanoparticles and nanotubes under the same conditions, the FΔF exhibited higher loading capacity (almost 68% W/W) than curcumin compared to other peptides.

    Furthermore, in the case of (IC50, 13 µM), this peptide group showed a higher parasite reproduction inhibition potential than pure curcumin in the P. falciparum-infected mice. The curcumin-containing dipeptide nanoparticles demonstrated a good system for drug delivery to the cells and inhibition of malaria parasite reproduction [49]. The study conducted by Kunwittaya et al. (2014) addressed the potential role of curcumin in parasite elimination and endothelial protection in case of cerebral malaria. According to the results, curcumin exhibited a high malaria growth inhibition level (IC50 ~10 µM) and reduced the endothelial apoptosis of brain vessels by 60%, while the pretreatment endothelial apoptosis was 79.6% [52]. In another study, edible curcumin nanoparticles were administrated to mice that infected by P. berghei. This treatment resulted in complete elimination of parasites from the patient's bodies and subsequently increased survival time of the infected mice from eight days to two months [51]. Furthermore, curcumin-chitosan nanoparticles (CCsNPs) enhanced the lifetime of mice which infected by P. yoelii. An in vitro experiment showed that CCsNPs could inhibit the growth of parasite through inducing the synthesis of hemozoin [50].

    A. castellanii is one of eight major protozoa causing severe keratitis in contact lens users. It can also cause skin lesions in patients with compromised immune systems [14]. Although medications against Acanthamoeba are commonly exploited, the complete eradication of infection seems to be a serious challenge when the parasite forms of the drug-resistant cysts. Therefore, pharmacotherapy of the Acanthamoeba or its cysts could emerge the new drug-resistant parasites [74].

    The study of El-Sayed et al. (2012) on the antiparasitic effect of ethanolic extract of C. longa against A. castellanii, two inhibition doses was determined in 48 h and 72 h with MIC of 1 g/mL and 100 mg/mL, respectively. This study showed that the ethanolic extract of C. longa had an inhibitory effect on the growth of the A. castellanii cysts with a MIC of 1 g and 100 mg/mL. The cysticidal effect of C. longa might be due to the curcumin, which is one of the major elements responsible for biological actions [54].

    Intestinal protozoa known as Entamoeba histolytica causes amoebiasis. As one of the main parasitic pathogens, this protozoan is estimated to infect 40–50 million individuals, among whom only 10% manifest the acute form of the disease leading to an annual mortality rate of 40000–100000 cases [16]. Metronidazole is an appropriate anti protozoan medication against the amoebic colitis; nevertheless, due to its fast absorption in case of oral infusion, it cannot be effective against the luminal trophozoites. Additionally, nearly 40–60% of the treatment is associated with drug-resistance of the parasite, so that elimination of the luminal parasites necessitates administration of paromomycin or diloxanide furoate for the patient [75]. Moreover, all of the drugs of this type are associated with some side effects such as nausea, headache, loss of appetite (anorexia), and metallic taste [2]. Therefore, researchers are still attempting to find new drugs with high therapeutic potential as well as low side effects.

    In a study in this regard, the researchers compared the amoebicidal capability of curcumin on E. hisolytica and its synergic effects with those of metronidazole. The best inhibitory effect of curcumin was achieved at concentration of 300 µM in 24 hours so that the parasite growth inhibition power was 65.5% and only 28.8% of the trophozoites could survive. Curcumin caused some morphological alterations in trophozoites, reduced parasite size, as well as reduced parasite uniformity. Such changes were examined using SEM microscope leading to the conclusion that the synergistic effect of curcumin and metronidazole would enhance the antiparasitic effects of these two medications together [55].

    Trichomoniasis is a sexually transmitted protozoan disease in human reproductive system which is caused by T. vaginalis. This disease is estimated to have an annual morbidity rate of 174 million cases worldwide. The drug-resistance engendered by metronidazole, which is the medication for T. vaginlis, necessitates the use of alternative drugs against this type of parasite [17].

    In another study, Watcher et al. (2014) addressed the role of curcumin derived from C. longa in the treatment of T. vaginalis. For this purpose, the strains with different sensitivities to metronidazole were exposed to curcumin treatment at concentrations of 400,200, and 800 µg/mL to compare with metronidazole. As a result, the best concentrations of curcumin with EC50 (73.0–105.8 µg/mL) and EC90 (64.9–216.3 µg/mL) at 3, 6, and 24 h showed that the concentration of 400 µg/mL could accomplish complete (100%) elimination of the parasites in 24 h [56]. Regarding the lower toxicity of curcumin at higher concentrations compared to metronidazole that is associated with systemic side effects in case of oral intake, and since the curcumin-containing drug (CurcumallR) is used topically for trichomoniasis treatment, it has been used with no toxic effect on the treatment of oral lesions [76].

    G. lambelia is a parasite with global distribution that infects nearly 280 million individuals annually. It is the most prevalent parasite in developed countries. In Africa, Asia, and Latin America, there are 200 million individuals with giardia infection, 500000 cases of which have clinical symptoms [19]. Metronidazole is one of the best medications for Giardia infection, yet it is associated with some side effects, such as nausea and metallic taste in mouth, and should not be taken with alcohol. Nitazoxanide (Alinia) is the liquid form of a drug prescribed for children, but it is also associated with side effects such as nausea, tympanites (meteorism), and yellow eyes [77]. Regarding the side effects of these drugs as well as their complications for pregnant women, specifically in the first trimester of pregnancy, it seems necessary to find and use some new medications for treating the Giardia infections.

    Gutierrez et al. (2017) showed the role of curcumin in the inhibition of Giardia. At 3 and 15 µM of curcumin, the cellular structure of Giardia was destroyed and then caused deformation of the parasite membrane, flagella, and abdominal wall. Based on the results of this study, tubulin is the main target of curcumin in destroying the Giardia lamblia trophozoite [57]. Additionally, the nanoparticles of silver, chitosan, and curcumin were also used for the treatment of giardiasis. As indicated by findings of this study, the use of these three types of nanoparticles combined together (nAg + Ncs + nCur) was effectively efficient compared to the separate use of them against Giardia, since the intestines and stool could be successfully cleaned and disposed of parasites while application of nAg + nCur for treatment of Giardia yielded a reduction of 91% in the parasites [58].

    T. gondii can afflict a wide range of warm-blooded animals including humans, livestock, and domestic animals. Clinically, this parasite causes some deficiencies in a growing embryo, which may lead to abortion in some cases, as well as opportunistic infections in immune-deficient patients and transplantation patients [20]. In acute forms of toxoplasmosis, two drugs known as pyrimethamine (Daraprim) and sulfadiazine are commonly used for medication. However, the pyrimethamine-induced side effects, such as bone marrow suppression and hepatotoxicity (liver toxicity), reveal the necessity of seeking to find new treatment alternatives for toxoplasmosis treatment purposes [78].

    Goo et al. (2015) reported that curcumin could inhibit the glyoxalase-1, thus plays an effective role in the anti-toxoplasmosis activity. By preventing the enzymatic activity of the recombinant TgGlo1, the inhibitory effect of curcumin resulted in the parasite proliferation in the T. gondii culture medium at IC50 38.3 ± 0.9 µM, leading to inhibition of the glyoxalase system metabolic pathway, which is necessary for the parasite to survive and live. These findings exhibited the role of curcumin against toxoplasmosis metabolic pathways [59].

    Neosporosis is caused by N. caninum and is a serious disease among cattle and dogs. This protozoon can cause abortion and stillbirth among cows [23]. As Qian W et al. (2015) showed in their study, curcumin was capable of preventing proliferation and growth of the parasites in human foreskin fibroblast cells infected by Neospora parasite at IC50 value of 1.1 ± 0.4 µM [22].

    C. parvum is one of the most important water-transmitted protozoans causing parasitic diseases, which is scattered throughout the world. It is transmitted through the oral-fecal route as well as contaminated water and foods. In patients with immune-deficiency, cryptosporidiosis can cause severe self-limiting diarrhea for two weeks, the treatment of which includes fluid consumption (for rehydration) and pain control [24]. Nitazoxanide is the drug recommended by FDA as the medication for treating diarrhea in immunocompetent patients, but its effectiveness among immunocompromised patients is still unknown [79]. The lack of an established treatment method for patients with immune deficiency, it seems necessary to seek new treatments.

    In Cryptosporidium-infected mice, in addition to the reduction in tissue lesions as well as some oocytes on the villi in both ileum and jejunum regions, curcumin could yield the increased TAC (total antioxidant capacity) and decreased MDA (malondialdehyde) in the infected mice [12].

    Parasites of this genus are capable of producing spores and releasing immature oocytes from the intestine's walls. In the poultry industry, E. tenella has caused health problems, decreased production efficiency, and increasingly elevated drug-resistance against antibiotics [27]. In one of the studies in this regard, different concentrations of curcumin were used to investigate the life cycle and morphology of the sporozoites. In this work, the curcumin concentrations of 100 and 200 µM reduced the sporozoite infection by 41.6% and 72.8%, respectively [61]. Additionally, daily C. longa-supplemented feeding of the broilers in a poultry farming led to reduction in the Eimeria-induced inflammatory reactions and intestinal damages [60]. In another study, the use of VAC (carvacrol, cinnamaldehyde, and capsicum oleoresin) edible compound yielded the improved immunity in the experimentally E. tenella-infected chickens [26].

    Trypanosoma is a flagellated protozoan member of the genus kinetoplastids. It is transmitted through biting or blood-feeding invertebrates and can cause some human diseases such as Chagas (caused by T. cruzi), which is mainly in Central and South America, and sleeping sickness (caused by T. brucei) mainly in Africa [80]. Pentamidine is a medication used in the primary stage of treatment for the sleeping sickness caused by T.b.gambiense infection, while other drugs (such as eflornithine, melarsoprol, suramin, and nifurtimox) are used for treating the African trypanosomiasis [81]. Also, benznidazole is the drug used for the treatment of Chagas disease, yet in case of acute form of the disease, nifurtimox is used as medication for treatment purposes [82]. Nevertheless, since the drugs used for treatment, including nifurtimox, has not been approved by FDA, it seems necessary to seek for new herbal medicines for treatment.

    Curcumin has the potential for obstruction, Ca2+-dependent NFATc1 duplication activities, induction of COX-2 and mPGES-1, production of PGE2 in excited ET-1 and parasite-infected myocytes, reduction of the level of prostaglandins of cardiomyocytes, as well as incurrence of disorders in PGE2/EP4 receptors [64]. In another study, examination of the morphological structure of the parasite using an electron microscope showed that the use of curcumin with IC50 value of 10.13 µM and dimetoxycurcumin (DMC) with IC50 value of 11.07 µM could result in cytoskeletal alterations in the parasite's epimastigote [29].

    Shistosomiasis is one of the most common parasitic diseases throughout the world, which is caused by a parasite called Schistosoma. There are about 207 million cases of this disease in 76 countries from around the world, mainly in developing countries, and meanwhile, 800 million individuals are exposed to the risk of infection [33].

    There are only a few drugs for the treatment of schistosomiasis, among which metrifonate is used for the urinary schistosomiasis and praziquantel is prescribed for all forms of schistosomiasis [83]. The emergence of drug resistance in this disease reveals the necessity of further studies aiming to find new medications.

    Hussein et al. (2017) conducted a study focusing on the potential role of C. longa, either dissolved lonely in olive oil or in combination with praziquantel, in treating the schistosomiasis mansoni (S. mansoni). According to the results, the use of C. longa combined with praziquantel led to the reduced worm burden and complete elimination of adult worms of S. mansoni in the studied mice, significantly reduced size of granuloma, a considerable improvement in worm-induced pathological damages in general, and a mild reduction in the inflammation among the mice [32]. Additionally, another study was conducted on the effect of curcumin on induction of apoptosis in the S. mansoni worms, the results of which indicated that curcumin caused DNA damages and fragmentation, increased expression of the SmCASP3/7 transcripts, and activation of Caspase 3 in male-female worm couples. Moreover, curcumin could increase the superoxide anion and activate the superoxide dismutase (SOD), which led to the oxidation of proteins in S. mansoni worms, indicating that induction of apoptotic-like events would increase the oxidative stress among the worms followed by their death [65]. Morias et al. (2013) showed that curcumin resulted in the separation of male and female S. mansoni worms, infertility of eggs, reduced oviposition, reduced lifetime of worms, and finally their death. Besides, it affected the expression of those genes that are involved in embryogenesis and oogenesis such as Notch and TGF-β. Curcumin affects the oviposition and growth of the parasite eggs by repressing the transcripts of these genes [66].

    The species of liver trematodes, such as F. hepatica and gigantica, can cause fasciolosis and are widely scattered throughout the world, mainly among the ruminants and among the sheep in particular. F. parasites infect the humans less than livestock. Both young and adult forms of these parasites can be treated by triclabendazole [34]. Ullah et al. (2017) showed that the use of curcumin at concentration of 60 µM resulted in a significantly reduced motility of the F. Gigantica worms (p < 0.05), disordered egg shedding, disrupted tegument, severely eroded spines in posterior region revealing the syncytium, as well as the significantly reduced activity of the GST enzyme at concentration of 60 µM (p < 0.01) [67].

    Moreover, the ovicidal properties of the curcumin-nisin nanoparticles against the Fasciola species were assessed. Accordingly, at the highest concentration of the curcumin-nisin nanoparticles, i.e., 5 mg/mL, the fasciola eggs had the lowest hatching rate (41.7%) compared to other groups and the control group, as well as the albendazole positive control group with hatching rate of 45.1%. Besides, the curcumin-nisin medication had no toxic effect on the mice's sperms and only killed the fasciola parasite eggs [68].

    The present review manifested the potential of curcumin as a common food seasoning with the hopes of increasing the knowledge of society about the various advantages of consumption of C. longa. Thus, further studies in formulations of curcumin and its derivatives could help to control, prevent, and treat the parasitic diseases.

    All authors declare no conflicts of interest in this review paper.

    [1] Croft SL, Sundar S, Fairlamb AH (2006) Drug resistance in leishmaniasis. Clin Microbiol Rev 19: 111–126.
    [2] Hayat F, Azam A, Shin D (2016) Recent progress on the discovery of antiamoebic agents. Bioorg Med Chem Lett 26: 5149–5159.
    [3] Oryan A, Akbari M (2016) Worldwide risk factors in leishmaniasis. Asian Pac J Trop Med 9: 925–932.
    [4] Aggarwal BB, Sundaram C, Malani N, et al. (2007) Curcumin: The Indian solid gold, In: The molecular targets and therapeutic uses of curcumin in health and disease, 1–75.
    [5] Agarwal A, Saxena PN (2018) Curcumin, a polyphenol from the Curcuma longa, prevents mercuric chloride-induced liver damage through reversal of oxidative stress and biochemical changes. AACR.
    [6] Aggarwal BB, Kumar A, Bharti AC (2003) Anticancer potential of curcumin: preclinical and clinical studies. Anticancer Res 23: 363–398.
    [7] Galasso V, Kovac B, Modelli A, et al. (2008) Spectroscopic and theoretical study of the electronic structure of curcumin and related fragment molecules. J Phys Chem A 112: 2331–2338.
    [8] Sun J, Zhao Y, Hu J (2013) Curcumin inhibits imiquimod-induced psoriasis-like inflammation by inhibiting IL-1beta and IL-6 production in mice. PloS one 8: e67078.
    [9] Aggarwal BB, Sung B (2009) Pharmacological basis for the role of curcumin in chronic diseases: An age-old spice with modern targets. Trends Pharmacol Sci 30: 85–94.
    [10] Strimpakos AS, Sharma RA (2008) Curcumin: Preventive and therapeutic properties in laboratory studies and clinical trials. Antioxid Redox Signal 10: 511–546.
    [11] Chen C, Johnston TD, Jeon H, et al. (2009) An in vitro study of liposomal curcumin: Stability, toxicity and biological activity in human lymphocytes and Epstein-Barr virus-transformed human B-cells. Int J Pharm 366: 133–139.
    [12] Asadpour M, Namazi F, Razavi SM, et al. (2018) Comparative efficacy of curcumin and paromomycin against Cryptosporidium parvum infection in a BALB/c model. Vet Parasitol 250: 7–14.
    [13] Feng J, Zhang L, Huang F, et al. (2018) Ready for malaria elimination: Zero indigenous case reported in the People's Republic of China. Malar J 17: 315.
    [14] Martinez A, Janitschke K (1985) Acanthamoeba, an opportunistic microorganism: A review. Infection 13: 251–256.
    [15] Baig AM (2015) Pathogenesis of amoebic encephalitis: Are the amoebae being credited to an "inside job" done by the host immune response? Acta Trop 148: 72–76.
    [16] Petri Jr W, Haque R, Lyerly D, et al. (2000) Estimating the impact of amebiasis on health. Parasitol Today 16: 320–321.
    [17] McClelland RS, Sangaré L, Hassan WM, et al. (2007) Infection with Trichomonas vaginalis increases the risk of HIV-1 acquisition. J Infect Dis 195: 698–702.
    [18] Minetti C, Chalmers RM, Beeching NJ, et al. (2016) Giardiasis. BMJ 40: 241–251.
    [19] Guy RA, Xiao C, Horgen PA (2004) Real-time PCR assay for detection and genotype differentiation of Giardia lamblia in stool specimens. J Clin Microbiol 42: 3317–3320.
    [20] Tenter AM, Heckeroth AR, Weiss LM (2000) Toxoplasma gondii: From animals to humans. Int J Parasitol 30: 1217–1258.
    [21] Cook AJC, Holliman R, Gilbert RE, et al. (2000) Sources of Toxoplasma infection in pregnant women: European multicentre case-control study Commentary: Congenital Toxoplasmosis—further thought for food. BMJ 321: 142–147.
    [22] Qian W, Wang H, Shan D, et al. (2015) Activity of several kinds of drugs against Neospora caninum. Parasitol Int 64: 597–602.
    [23] Dubey J, Lindsay D (1996) A review of Neospora caninum and neosporosis. Vet Parasitol 67: 1–59.
    [24] Checkley W, Epstein LD, Gilman RH, et al. (1998) Effects of Cryptosporidium parvum infection in Peruvian children: Growth faltering and subsequent catch-up growth. Am J Epidemiol 148: 497–506.
    [25] Kramer MH, Herwaldt BL, Craun GF, et al. (1996) Surveillance for waterborne-disease outbreaks—United States, 1993–1994. MMWR CDC Surveill Summ 12: 1–33.
    [26] Lee SH, Lillehoj HS, Jang SI, et al. (2011) Effects of dietary supplementation with phytonutrients on vaccine-stimulated immunity against infection with Eimeria tenella. Vet Parasitol 181: 97–105.
    [27] Giannenas I, Florou-Paneri P, Papazahariadou M, et al. (2003) Effect of dietary supplementation with oregano essential oil on performance of broilers after experimental infection with Eimeria tenella. Arch Anim Nutr 57: 99–106.
    [28] Novaes RD, Sartini MVP, Rodrigues JPF, et al. (2016) Curcumin enhances the anti-Trypanosoma cruzi activity of benznidazole-based chemotherapy in acute experimental Chagas disease. Antimicrob Agents Chemother 60: 3355–3364.
    [29] Sueth-Santiago V, de BB Moraes J, Alves ESS, et al. (2016) The effectiveness of natural diarylheptanoids against Trypanosoma cruzi: Cytotoxicity, ultrastructural alterations and molecular modeling studies. PLoS One 11: e0162926.
    [30] Baker J (1995) The subspecific taxonomy of Trypanosoma brucei. Parasite 2: 3–12.
    [31] Deborggraeve S, Koffi M, Jamonneau V, et al. (2008) Molecular analysis of archived blood slides reveals an atypical human Trypanosoma infection. Diagn Micr Infect Dis 61: 428–433.
    [32] Hussein A, Rashed S, El Hayawan I, et al. (2017) Evaluation of the Anti-schistosomal Effects of Turmeric (Curcuma longa) Versus Praziquantel in Schistosoma mansoni Infected Mice. Iran J Parasitol 12: 587–596.
    [33] Oluwole AS, Adeniran AA, Mogaji HO, et al. (2018) Prevalence, intensity and spatial co-distribution of schistosomiasis and soil transmitted helminths infections in Ogun state, Nigeria. Parasitol Open 4.
    [34] Sarkari B, Khabisi SA (2017) Immunodiagnosis of Human Fascioliasis: An Update of Concepts and Performances of the Serological Assays. J Clin Diagn Res 11: OE05–OE10.
    [35] Bauri R, Tigga MN, Kullu SS (2015) A review on use of medicinal plants to control parasites. Indian J Nat Prod Resour 6: 268–277.
    [36] Wink M (2012) Medicinal plants: A source of anti-parasitic secondary metabolites. Molecules 17: 12771–12791.
    [37] Hatcher H, Planalp R, Cho J, et al. (2008) Curcumin: From ancient medicine to current clinical trials. Cell Mol Life Sci 65: 1631–1652.
    [38] Mohammad BA (2011) Antileishmanial effects of traditional herbal extracts against cutaneous leishmaniosis in vivo. Adv Environ Biol 5: 3188–3195.
    [39] Hosseini A, Jaffary F, Asghari GR, et al. (2012) In Vitro Effects of Turmeric and Licorice Total Extracts on L. Major Promastigotes. J Isfahan Med Sch 29: 1–11.
    [40] Khanra S, Kumar YP, Dash J, et al. (2018) In vitro screening of known drugs identified by scaffold hopping techniques shows promising leishmanicidal activity for suramin and netilmicin. BMC Res Notes 11: 319.
    [41] Amaral ACF, Gomes LA, Silva JRdA, et al. (2014) Liposomal formulation of turmerone-rich hexane fractions from Curcuma longa enhances their antileishmanial activity. BioMed Res Int 2014: 694934.
    [42] Fouladvand M, Barazesh A, Tahmasebi R (2013) Evaluation of in vitro antileishmanial activity of curcumin and its derivatives "gallium curcumin, indium curcumin and diacethyle curcumin". Eur Rev Med Pharmacol Sci 17: 3306–3308.
    [43] Tiwari B, Pahuja R, Kumar P, et al. (2017) Nanotized curcumin and miltefosine, a potential combination for treatment of experimental visceral leishmaniasis. Antimicrob Agents Chemother 61: e01169-16.
    [44] Busari ZA, Dauda KA, Morenikeji OA, et al. (2017) Antiplasmodial Activity and Toxicological Assessment of Curcumin PLGA-Encapsulated Nanoparticles. Front Pharmacol 8: 622.
    [45] Donipati P, Hara Sreeramulu S (2015) In vitro anti-malarial activity of Rhizome extracts of Curcuma species. Int J Pharm Bio Sci 5: 488–494.
    [46] Vathsala PG, Dende C, Nagaraj VA, et al. (2012) Curcumin-arteether combination therapy of Plasmodium berghei-infected mice prevents recrudescence through immunomodulation. PLoS One 7: e29442.
    [47] Singh DB, Dwivedi S (2016) Structural insight into binding mode of inhibitor with SAHH of Plasmodium and human: interaction of curcumin with anti-malarial drug targets. J Chem Biol 9: 107–120.
    [48] Dende C, Meena J, Nagarajan P, et al. (2015) Simultaneously targeting inflammatory response and parasite sequestration in brain to treat Experimental Cerebral Malaria. Sci Rep 5: 12671.
    [49] Alam S, Panda JJ, Mukherjee TK, et al. (2016) Short peptide based nanotubes capable of effective curcumin delivery for treating drug resistant malaria. J Nanobiotechnol 14: 26.
    [50] Akhtar F, Rizvi MMA, Kar SK (2012) Oral delivery of curcumin bound to chitosan nanoparticles cured Plasmodium yoelii infected mice. Biotechnol Adv 30: 310–320.
    [51] Ghosh A, Banerjee T, Bhandary S, et al. (2014) Formulation of nanotized curcumin and demonstration of its antimalarial efficacy. Int J Nanomedicine 9: 5373–5387.
    [52] Kunwittaya S, Treeratanapiboon L, Srisarin A, et al. (2014) In vitro study of parasite elimination and endothelial protection by curcumin: Adjunctive therapy for cerebral malaria. EXCLI J 13: 287–299.
    [53] Dohutia C, Chetia D, Gogoi K, et al. (2017) Design, in silico and in vitro evaluation of curcumin analogues against Plasmodium falciparum. Exp Parasitol 175: 51–58.
    [54] El-Sayed NM, Ismail KA, Ahmed SAEG, et al. (2012) In vitro amoebicidal activity of ethanol extracts of Arachis hypogaea L., Curcuma longa L. and Pancratium maritimum L. on Acanthamoeba castellanii cysts. Parasitol Res 110: 1985–1992.
    [55] Rangel‐Castañeda IA, Hernández‐Hernández JM, Pérez‐Rangel A, et al. (2018) Amoebicidal activity of curcumin on Entamoeba histolytica trophozoites. J Pharm Pharmacol 70: 426–433.
    [56] Wachter B, Syrowatka M, Obwaller A, et al. (2014) In vitro efficacy of curcumin on Trichomonas vaginalis. Wien Klin Wochenschr 126: 32–36.
    [57] Gutiérrez-Gutiérrez F, Palomo-Ligas L, Hernández-Hernández JM, et al. (2017) Curcumin alters the cytoskeleton and microtubule organization on trophozoites of Giardia lamblia. Acta Trop 172: 113–121.
    [58] Said D, Elsamad L, Gohar Y (2012) Validity of silver, chitosan, and curcumin nanoparticles as anti-Giardia agents. Parasitol Res 111: 545–554.
    [59] Goo YK, Yamagishi J, Ueno A, et al. (2015) Characterization of Toxoplasma gondii glyoxalase 1 and evaluation of inhibitory effects of curcumin on the enzyme and parasite cultures. Parasites Vectors 8: 654.
    [60] Kim DK, Lillehoj HS, Lee SH, et al. (2013) Dietary Curcuma longa enhances resistance against Eimeria maxima and Eimeria tenella infections in chickens. Poultry Sci 92: 2635–2643.
    [61] Khalafalla RE, Müller U, Shahiduzzaman M, et al. (2011) Effects of curcumin (diferuloylmethane) on Eimeria tenella sporozoites in vitro. Parasit Res 108: 879–886.
    [62] Burt S, Tersteeg-Zijderveld M, Jongerius-Gortemaker B, et al. (2013) In vitro inhibition of Eimeria tenella invasion of epithelial cells by phytochemicals. Vet Parasitol 191: 374–378.
    [63] Nagajyothi F, Zhao D, Weiss LM, et al. (2012) Curcumin treatment provides protection against Trypanosoma cruzi infection. Parasitol Res 110: 2491–2499.
    [64] Hernández M, Wicz S, Corral RS (2016) Cardioprotective actions of curcumin on the pathogenic NFAT/COX-2/prostaglandin E2 pathway induced during Trypanosoma cruzi infection. Phytomedicine 23: 1392–1400.
    [65] de Paula Aguiar D, Moscardini MBM, Morais ER, et al. (2016) Curcumin Generates Oxidative Stress and Induces Apoptosis in Adult Schistosoma mansoni Worms. PLoS One 11: e0167135.
    [66] Morais ER, Oliveira KC, Magalhães LG, et al. (2013) Effects of curcumin on the parasite Schistosoma mansoni: A transcriptomic approach. Mol Biochem Parasit 187: 91–97.
    [67] Ullah R, Rehman A, Zafeer MF, et al. (2017) Anthelmintic potential of thymoquinone and curcumin on Fasciola gigantica. PLoS One 12: e0171267.
    [68] Oyeyemi O, Adegbeyeni O, Oyeyemi I, et al. (2018) In vitro ovicidal activity of poly lactic acid curcumin-nisin co-entrapped nanoparticle against Fasciola spp. eggs and its reproductive toxicity. J Basic Clin Physiol Pharmacol 29: 73–79.
    [69] Desta A, Shiferaw S, Kassa A, et al. (2005) Module on Leishmaniasis for the Ethiopian Health Center Team. Debub University, Ethiopia.
    [70] Ameen M (2007) Cutaneous leishmaniasis: Therapeutic strategies and future directions. Expert Opin Pharmacother 8: 2689–2699.
    [71] Lehane AM, Kirk K (2010) Efflux of a range of antimalarial drugs and "chloroquine resistance reversers" from the digestive vacuole in malaria parasites with mutant PfCRT. Mol Microbiol 77: 1039–1051.
    [72] Padmanaban G, Nagaraj VA, Rangarajan PN (2012) Artemisinin-based combination with curcumin adds a new dimension to malaria therapy. Curr Sci 102: 704–711.
    [73] Cui L, Miao J, Cui L (2007) Cytotoxic effect of curcumin on malaria parasite Plasmodium falciparum: Inhibition of histone acetylation and generation of reactive oxygen species. Antimicrob Agents Chemother 51: 488–494.
    [74] Obeid WN, de Araújo R, Vieira LA, et al. (2003) Acanthamoeba bilateral keratiti—Case report. Rev Bras Oftalmol 66: 876–880.
    [75] Farthing MJ (2006) Treatment options for the eradication of intestinal protozoa. Nat Rev Gastroenterol Hepatol 3: 436-445.
    [76] Meidan I, Sellam G, Simaan S, et al. (2013) Topical curcumin for the prevention of oral mucositis in pediatric patients: Case series. Altern Ther Health Med 19: 21-24.
    [77] Granados CE, Reveiz L, Uribe LG, et al. (2012) Drugs for treating giardiasis. Cochrane database of systematic reviews 12: CD007787.
    [78] Gilbert R, Gras L, Wallon M, et al. (2001) Effect of prenatal treatment on mother to child transmission of Toxoplasma gondii: Retrospective cohort study of 554 mother-child pairs in Lyon, France. Int J Epidemiol 30: 1303–1308.
    [79] Mofenson LM, Brady MT, Danner SP, et al. (2009) Guidelines for the prevention and treatment of opportunistic infections among HIV-exposed and HIV-infected children: Recommendations from CDC, the National Institutes of Health, the HIV Medicine Association of the Infectious Diseases Society of America, the Pediatric Infectious Diseases Society, and the American Academy of Pediatrics. MMWR Recomm Rep 58: 1–198.
    [80] Stephen LE (1986) Trypanosomiasis: A veterinary perspective. Pergamon Press, Oxford.
    [81] Babokhov P, Sanyaolu AO, Oyibo WA, et al. (2013) A current analysis of chemotherapy strategies for the treatment of human African trypanosomiasis. Pathog Global Health 107: 242–252.
    [82] Kowalska A, Kowalski P, Torres MÁT (2011) Chagas disease–american trypanosomiasis. Pol Ann Med 18: 156–167.
    [83] Zanolla D, Perissutti B, Passerini N, et al. (2018) Milling and comilling Praziquantel at cryogenic and room temperatures: Assessment of the process-induced effects on drug properties. J Pharmaceut Biomed 153: 82–89.
  • This article has been cited by:

    1. Muslimah Alsulami, Rewaida Abdel-Gaber, Cell therapy as a new approach on hepatic fibrosis of murine model of Schistosoma mansoni-infection, 2021, 66, 1230-2821, 136, 10.1007/s11686-020-00267-2
    2. Aishah E. Albalawi, Abdullah D. Alanazi, Iraj Sharifi, Fatemeh Ezzatkhah, A Systematic Review of Curcumin and its Derivatives as Valuable Sources of Antileishmanial Agents, 2021, 1230-2821, 10.1007/s11686-021-00351-1
    3. Soo-In Sohn, Arumugam Priya, Boopathi Balasubramaniam, Pandiyan Muthuramalingam, Chandran Sivasankar, Anthonymuthu Selvaraj, Alaguvel Valliammai, Ravi Jothi, Subramani Pandian, Biomedical Applications and Bioavailability of Curcumin—An Updated Overview, 2021, 13, 1999-4923, 2102, 10.3390/pharmaceutics13122102
    4. Ivona Mladineo, Željka Trumbić, Adrián Ormad-García, Oswaldo Palenzuela, Ariadna Sitjà-Bobadilla, Simona Manuguerra, Cristobal Espinosa Ruiz, Concetta Maria Messina, In Vitro Testing of Alternative Synthetic and Natural Antiparasitic Compounds against the Monogenean Sparicotyle chrysophrii, 2021, 10, 2076-0817, 980, 10.3390/pathogens10080980
    5. Dennis R.A. Mans, Priscilla Friperson, Meryll Djotaroeno, Jennifer Pawirodihardjo, 2022, Chapter 13, 978-1-80356-020-5, 10.5772/intechopen.97751
  • Reader Comments
  • © 2018 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(11693) PDF downloads(362) Cited by(4)

Figures and Tables

Figures(1)  /  Tables(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog