Please cite this paper as:
Honorio, R.I., Dias, B.K.M., Przyborski, J.M. and Garcia, C.R. 2022. Melatonin as a microenvironmental cue for parasite development inside the host. Melatonin Research. 5, 1 (Apr. 2022), 84-100. DOI:https://doi.org/https://doi.org/10.32794/mr112500122.
Review
Melatonin as a microenvironmental cue for parasite development inside the host
Rute Isabel Honorio1, Barbara Karina Menezes Dias1,2, Jude Marek Przyborski3 and Celia Regina Garcia1
1Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil.
2Department of Parasitology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil.
3Department of Biochemistry and Molecular Biology (BiMoBi), Interdisciplinary Research Center (iFZ), Justus-Liebig-University, Giessen, Germany.
*Correspondence: cgarcia@usp.br, Tel:5511-3091-1505
Running title: Melatonin interactions in the host-parasite relationship
Received: February 13, 2022, Accepted: April 8, 2022
ABSTRACT
Throughout the evolutionary process, parasites have acquired characteristics that function as survival mechanisms. It has been reported that melatonin, a molecule present in virtually all living organisms, has several roles in parasite biology such as preventing tissue damage, regulating gene expression and inflammatory processes, and acting as a free radical scavenger. Additionally, melatonin produced by the hosts accelerates the intra-erythrocytic cycle of the human malaria parasite Plasmodium falciparum and the rodent malaria parasite P. chabaudi, respectively. These findings have recently led to an increased research enthusiasm to find how melatonin influences the biological cycle of parasites. Therefore, this review aims to gather and analyze the potential relationships of host produced melatonin with the parasites Plasmodium sp., Trypanosoma cruzi, Leishmania spp., Toxoplasma gondii, Schistosoma mansoni, Opisthorchis viverrini, and Entamoeba histolytica, respectively.
Key words: melatonin, parasites, pathway signaling, cAMP, analogs, malaria, receptor
___________________________________________________________________________________________________________
1. INTRODUCTION
Melatonin is an indolamine molecule present in almost all living organisms, from bacteria and fungi to mammals (1). The free-radical scavenger and immunomodulatory activity of melatonin have been reported over the years in a variety of biologic conditions (2, 3). The actions of melatonin as an antioxidant and modulator of immune response to protect tissue damage have been evidenced by many in vitro and in vivo studies. For example, Leite et al. (4) reported that melatonin treatment before intestinal ischemia-reperfusion protected the acute lung injury with the reduced oxidative stress. In diabetic rat model with the increased malondialdehyde and superoxide dismutase levels, indicative of oxidative stress, melatonin treatment was able to reverse these parameters (5). Besides its antioxidant and anti-inflammatory effects for the treatment of cancer, the influence of melatonin on apoptosis and cell proliferation processes has been studied in vitro with promising results (6-8). Clinically, the use of melatonin for cancer treatments can improve the patient’s outcomes concerning remission, survival and reduced side effects of the chemotherapy (9). The use of melatonin to treat respiratory viral infections also show beneficial effects. For example, melatonin treatment can reduce the symptoms and mortalities of SARS-CoV-2 infected patients (10-12)
Besides the free-radical scavenger activity, there are two melatonin receptors, MT1 and MT2, which are G-protein coupled receptors (13, 14). In mammals, the downstream signaling of melatonin involves a decrease in cyclic adenosine monophosphate (cAMP) levels (15).
Melatonin was discovered in 1958 in the bovine pineal gland (16). The pineal gland is the main source of circulating melatonin in mammals and is released into the bloodstream and cerebrospinal fluid in a light/dark circadian rhythm (17-19). The presence of melatonin synthetic pathway in other organs strongly indicates that this molecule can be produced in other parts of the body (20-22). Indeed, it has been confirmed that melatonin is produced in mitochondria, therefore, virtually every cell or system including the immune system, platelets, the gastrointestinal tract also can synthesize melatonin (17, 23-27).
As to the melatonin biosynthesis in vertebrates, tryptophan is the precursor. Throughout the processes of hydroxylation, decarboxylation, acetylation, and methylation in order, tryptophan is converted to melatonin. The acetylation step is catalyzed by the aralkylamine N-acetyltransferase (AANAT) which is the rate-limiting enzyme for melatonin synthesis and its activity is regulated by the light/dark cycle (28). The light intensity detected by the retina is the neuronal signal via the retinohypothalamic tract transmitted to the suprachiasmatic nucleus of the hypothalamus. This leads to the alteration of norepinephrine release into the pineal perivascular space of the pineal gland (28, 29). Norepinephrine then acts on β1-adrenergic receptors leading to activation of downstream signaling via adenylyl cyclase, causing an increase in intracellular calcium (Ca2+) concentration, phosphokinase C activity (30, 31), and a rise in cAMP concentration in the pinealocytes (30, 32–34). cAMP is responsible for modulating AANAT activity by causing an increase in AANAT affinity for arylalkilamines (28, 35–37).
2. MELATONIN AND PLASMODIUM
Malaria is a severe disease caused by parasites of the Plasmodium genus and was estimated to be responsible for the infection of 229 million people and 409 thousand deaths in 2019 globally (38). Among Plasmodium species, P. falciparum, P. ovale, P. malariae, P. vivax, and P. knowlesi can infect humans (39, 40).
The infection of the host occurs during the feeding process of the female infected mosquito of the Anopheles genus, which injects sporozoites into the host bloodstream. The sporozoites migrate and establish the infection in the liver. In the hepatocytes, Plasmodium parasites undergo a series of multiplication and develop into merozoites, which are released into the bloodstream. Their intra-erythrocytic cycle starts when merozoites invade the host erythrocytes. During the erythrocytic cycle, Plasmodium parasites undergo three well-characterized stages: ring, trophozoite, and schizont, the last one forming merozoites, which are released into the bloodstream from the infected red blood cell (RBC). Merozoites can invade new erythrocytes and continue the asexual intra-erythrocytic cycle. A few parasites are committed to generating the gametocyte forms microgamete and macrogamete; the sexual form of the parasite that infects the mosquito vector. During the feeding process, a female mosquito from the Anopheles genus ingests the sexual forms of the parasites, initiating the sexual cycle in the mosquito (41, 42).
The duration of the intra-erythrocytic cycle of Plasmodium parasites is generally a multiple of 24 hours, resembling circadian rhythms. Moreover, some Plasmodium species development is synchronized with the host rhythms, a process characterized by a release peak of merozoites in the bloodstream (43, 44). Interestingly, Trager and Jansen (45) observed that P. falciparum loses the synchrony when maintained in culture. Among the Plasmodium species, only the human parasite P. falciparum have a synchronized intraerythrocytic cycle and the rodent parasite P. chabaudi (46), whereas P. yoelli and P. berghei do not exhibit such feature. (47).
Boyd et al. (48) investigated the influence of the host circadian rhythm in the chicken parasite P. cathemerium. They found that the inversion of female canaries' light/dark cycle is followed by an inversion of the merozoite release peak of the parasite. David et al. (49) elegantly showed that it was possible to invert the peak of schizogony of P. chabaudi by artificially inverting the host light/dark cycle. The pineal melatonin production is also associated with the light/dark cycle in vertebrates. From this point of view, the relationship between melatonin and Plasmodium sp. has drawn great attention of researchers.
To investigate whether melatonin could influence P. chabaudi intraerythrocytic cycle, Hotta et al. (46) evaluated the effect of pinealectomy on P. chabaudi-infected rats. Pinealectomy disrupted the synchrony of P. chabaudi and the effect was reverted upon melatonin treatment. The results showed that melatonin triggered an increase in cytosolic Ca2+ concentration (46, 50) and pre-treatment with a melatonin-receptor antagonist, luzindole, or with the phospholipase-C (PLC) inhibitor, U73122, prevented these changes. These data indicate that melatonin acts through a receptor and the release of intracellular calcium is mediated by PLC.
Beraldo and Garcia (51) showed that the incubation of parasites with melatonin precursors N-acetylserotonin, serotonin and tryptamine exerts a synchronizing effect and increases the cytosolic concentration of Ca2+ in P. falciparum in in vitro experiments (51, 52). Also, N1-acetyl-N2-formyl-5-methoxykynuramine, a melatonin metabolite, causes an increase in cytosolic Ca2+ concentration in vitro and influences the asexual cycle of P. falciparum and P. chabaudi (53). On the other hand, P. yoelii and P. berghei, parasites known for having an asynchronous cycle in the host, are neither influenced by melatonin in terms of synchronization nor change cytosolic Ca2+ concentration (47).
To assess the influence of melatonin related compounds on the P. falciparum asexual cycle, Schuck and collaborators (54) found that these melatonin-related compounds were able to reduce the increased parasitemia caused by melatonin and three of these compounds to achieve this effect were only required micromolar concentration in in vitro condition. Dias et al. (55) also treated the parasites with synthetic indole-like compounds, Triptiofen or Melatotosil. The outcome was interesting. Triptiofen was able to stop the parasite cell cycle and did not induce an increase in cytosolic Ca2+ concentration while Melatotosil induced an increase in cytosolic Ca2+ concentration and increased parasitemia, an effect similar to that when P. falciparum was treated with melatonin. These results open the way to explore indole-like compounds as a treatment for malaria caused by parasites that are influenced by melatonin.
Using isolated P. falciparum at the trophozoite stage, Beraldo et al. (56), observed that melatonin treatment, as expected, increased cytosolic Ca2+ concentration. To investigate the source of Ca2+, they incubated the parasites with melatonin in the presence and absence of extracellular calcium. In both situations, the increase in cytosolic Ca2+ concentration was similar. This result points out that melatonin triggers a rise in Ca2+ concentration by using an internal source.
Alves et al. (57) studied whether melatonin treatment could increase cytosolic Ca2+ concentration through activation of inositol 1,4,5-trisphosphate (IP3) receptor in the sarcoplasmic reticulum of P. falciparum. They used a labelled PLC-substrate, [3H-myo] inositol, and observed that melatonin treatment increased the metabolization of [3H-myo] inositol and increased cytosolic Ca2+ concentration. Using caged IP3, a way to control the intracellular IP3 concentration, the authors observed an increase in cytosolic Ca2+ concentration after induction of IP3 release. To elucidate whether melatonin and IP3 act in the same route to increase cytosolic Ca2+ concentration, P. falciparum was treated with melatonin followed by induction of cytosolic IP3 release, and the characteristic IP3-induced increase in cytosolic Ca2+ concentration was not observed. Thus, the downstream signaling of melatonin involves the activation of PLC and IP3 formation, which acts on the putative IP3 receptor at the sarcoplasmic reticulum, leading to the release of Ca2+ in the cytosol (57–59). Although IP3-induced Ca2+ release was demonstrated to be important, the IP3 receptor in Plasmodium is not yet identified. Using affinity chromatography and bioinformatic analysis, Alves et al. (57) identified the multi-drug resistance 1 (MDR1) transporter as a possible IP3 receptor (60). Still, more research is needed to confirm this hypothesis.
To elucidate whether the protein kinase A (PKA) would be involved in the intracellular signaling activation induced by melatonin in Plasmodium, Beraldo et al. (56) started an investigation with this kinase. To do so, they tested the influence of the cAMP analog, 6-Bz-cAMP, and the PKA inhibitor, PKI, in asynchronous P. falciparum cultures after melatonin treatment. Melatonin treatment caused an increase in parasitemia and also accelerated schizont formation. Treatment with 6-Bz-cAMP had the similar effect as melatonin, which was the increase of the schizont stage, while treatment with PKI prevented it (56). This is strong evidence that melatonin triggers downstream signaling involving cAMP and PKA.
The mitogen-activated protein kinase (MAPK) is a family of proteins that are involved in response to external stimuli in eukaryotic cells and are present in diverse organisms such as plants, mammals, and unicellular organisms (61). The regulation of MAPK activation involves a phosphorylation cascade, starting with the MAPK kinase kinase (MAPKKK) phosphorylating MAPK kinase (MAPKK), which phosphorylates MAPK (62, 63). Using BLAST analysis, Doring et al. (64) identified the protein kinase 7 (PfPK7), a MAPKK related protein in the P. falciparum genome, and the result was quite interesting: the C terminal is highly similar to MAPKK, and the N terminal to a fungal PKA. Using P. falciparum expressing a non-functional PfPK7, the authors (64) also observed that MAPKK and PKA inhibitors showed no effect in this strain, indicating that the classical MAPKKK-MAPKK-MAPK phosphorylation cascade did not occur in P. falciparum.
In 2020, Dorin-Semblat et al. (65) observed that the P. falciparum PK7 knockout (PfPK7-) strain had a lower parasite growth. Moreover, using PfPK7-, Koyama et al. (66) identified that this kinase was involved in the downstream signaling activated by melatonin. Treatment of PfPK7- parasites did not induce the characteristic synchronization exerted by melatonin in the wild-type strain, indicating that this kinase is essential for synchronization caused by melatonin. In addition, Koyama et al. (66) evaluated whether melatonin treatment would be able to affect the expression of genes related to the ubiquitin-proteasome system. A 5 hour-melatonin treatment in P. falciparum trophozoite stage (wild-type and PfPK7- strain) upregulated 13 of 14 studied genes in the wild-type strain and, interestingly, this effect was not present in PfPK7- parasites. Another kinase involved in the downstream response to melatonin in P. falciparum is the eukaryotic initiation factor 2 (eIF2) kinase 1 (PfeIK1). Melatonin cannot induce an increase in parasitemia in P. falciparum lacking PfeIK1, indicating that this kinase might be important for the synchronization effect exerted by melatonin in vivo (55). The eIF2α kinases family is responsible for phosphorylating the α subunit of eIF2, leading to the repression of the general translation of proteins (67).
As PKA is a kinase involved in melatonin downstream signaling in P. falciparum, Lima et al. (68) observed that treatment with 6-Bnz-cAMP, a PKA activator, caused an increase in levels of the transcription factor PfNF-YB in the trophozoite stage, and that melatonin treatment increases the ubiquitination of this transcription factor. These results indicate that melatonin treatment regulates PfNF-YB levels via ubiquitination through PKA and the second messenger cAMP. The transcription factor NF-YB binds to a CCAAT-box DNA sequence in the genome and modulates the expression of several genes in mammals (69–71). Although P. falciparum also expresses NF-YB (72), it is still unknown which genes are regulated by this particular transcription factor in this parasite.
A recent study has shown the rhythmic expression of genes in P. falciparum and P. chabaudi, suggesting that the parasites possess an intrinsic clock responsible for their rhythms (43). Considering the results of gene expression regulation induced by melatonin, Lima et al. (73) performed RNA-seq analysis of in vitro P. falciparum in the trophozoite stage after melatonin treatment and 38 genes were differentially expressed. These genes include the regulator of initiation factor 2 (eIF2), ubiquitin-40S ribosomal protein, protein tyrosine phosphatase PRL, EMP1 traffic protein PTP4, heterochromatin protein 1 (HP1), and DNA repair endonuclease (73). The results suggest that melatonin plays a key role to modulate the cell cycle of Plasmodium within the host through gene regulation. Treatment with melatonin modulates the expression of Mitochondrial Fission Protein 1 (FIS1) and the GTPases Dynamin 1 (DYN1) and Dynamin 2 (DYN2) genes, that encode proteins related to mitochondrial fission (74). There is no alteration in the expression of FIS1, DYN1, and DYN2 in the P. falciparum PfPK7- strain, indicating that PfPK7 is a kinase involved in modulating mitochondrial-fission gene expression (74). FIS1 is a protein involved in the fragmentation of the mitochondrial network and its action depends on the GTPases DYN1 and DYN2 in P. falciparum (74, 74). Corroborating with this idea, PfPK7- parasites have a slower multiplication rate due to the lower production of merozoites (65). Therefore, these data indicate that the mitochondrial fission step is very important to the generation of merozoites, the last intra-erythrocytic stage and that PfPK7 participates in this process. In addition, an increase in the michorchidia (MORC) protein levels under melatonin treatment in intra-erythrocytic stages of P. falciparum has been observed (76) and this effect is absent also in the PfPK7- strain. MORC is a protein found in plants and animals identified as a gene regulator by mediating chromatin remodeling, gene silencing, and epigenetic regulation (77, 78). P. falciparum expresses this protein - PfMORC - in its nucleus, but its function is not yet fully understood (61, 64). This information warrants further studies to identify the function of PfMORC in the Plasmodium intra-erythrocytic stages.
Although melatonin can activate several signaling pathways in Plasmodium, its receptor in this parasite is yet to be discovered. Melatonin receptors in humans are G protein-coupled receptors (GPCR), known for their serpentine characteristic with seven transmembrane domains (80, 81). Using in situ analysis, Madeira et al. (82) identified four serpentine receptors candidates in the P. falciparum genome, namely: PfSR1, PfSR10, PfSR12, and PfSR25.
In addition to melatonin-mediated activation of internal signaling cascades in Plasmodium sp., this molecule can also modulate the course of its infection in the host. By treating P. yoelii-infected mice with melatonin, Guha et al. (83) demonstrated a significant reduction in liver mitochondrial apoptosis and liver damage. The influence of melatonin in cerebral malaria has also been studied in P. berghei infected mice (84). This treatment prevented cerebral edema and impairment of the blood-brain barrier. Oral melatonin (3 mg) was reported to increase Plasmodium detection in the peripheral blood of patients (85). This result corroborates well with the result of Beraldo et al. (51), i.e., a significant increase in the schizonts forms was found when P. falciparum culture was treated with melatonin for 24 hours. The potential mechanisms of host melatonin on the Plasmodium falciparum life cycle are illustrated in figure 1.
Fig.1: Schematic representation of the effects of melatonin in Plasmodium falciparum.
Mel – melatonin; RBC – red blood cell; PVM – parasitophorous vacuole membrane; PV – parasitophorous vacuole; PM – parasite membrane; Created in BioRender.com.
3. MELATONIN AND TRYPANOSOMA CRUZI
Chagas disease is caused by Trypanosoma cruzi (T. cruzi) infection, with an estimation of 30,000 cases per year in the Americas and 12,000 deaths per year (86). The parasite cycle depends on the human host and the transmitting vector, which is an insect from the Triatominae family. The trypomastigotes of the parasite released during the blood sucking of the vector are transported to the wound or mucosa where they invade into the local cells. Inside the cells, the parasites develop to the stage of amastigotes and trypomastigotes, with the last being released in the bloodstream. From the blood, the trypomastigotes either initiate another cycle of cell invasion or are ingested by the vector (87).
The acute phase of infection is characterized by the presence of parasites in the bloodstream and can be asymptomatic. However, the chronic phase of Chagas disease is characterized by the presence of the parasite in tissues such as the heart and intestine (88). Although the etiology of the complications caused by Chagas disease is not fully understood, evidence shows that the immune system response plays a role in it (89, 90). Therefore, understanding the immune response against T. cruzi infection is essential to modulate the innate immune system against the parasite.
Santello et al. (91, 92) observed that melatonin-treatment concomitant with or before T. cruzi infection led to a decrease in parasitemia. In addition to acting as a free radical scavenger, melatonin treatment also influences the activity of the immune system with an increase in TNF-α, IFN-γ, IL-12, IL-2, a decrease in IL-4 levels, induction of leukocyte production, and suppression of Th2 response in Chagas disease (91–95). The main hypothesis of the decreased parasitemia levels upon melatonin treatment relies on the immunoregulatory effects of melatonin during the infection (91–95). For example, orchiectomy combined melatonin treatment had a synergistic effect on leukocyte count and IL-2 and IL-12 production (94). Melatonin combined with dehydroepiandrosterone (DHEA) treatment on acute-T. cruzi infected rats led to an increase in peritoneal macrophages and TNF-α levels and a decrease in parasitemia (95). The middle-aged rats infected with T. cruzi treated with melatonin caused the reduction of the steroidal hormones of corticosterone, 11-dehydrocorticosterone, cortisol, cortisone, aldosterone, and progesterone levels (96). The results indicate a close relationship between melatonin treatment and hormones in the progression of Chagas disease.
To explore the action of melatonin as a free radical scavenger in T. cruzi infection, Brazão et al. (97) observed that this indoleamine can decrease the production of superoxide (O2-) in the thymus of infected animals and increase superoxide dismutase (SOD) activity, a natural antioxidant (97, 98). The association of melatonin with zinc, an antioxidant and anti-inflammatory agent, has been studied and its crosstalk neutralizes immune dysregulation induced by the parasites in the chronic stage (99, 100). Melatonin treatment diminished the burden of amastigotes in heart tissue leading to less cardiac damage during the acute phase (92). The beneficial effects of melatonin can also be observed in the chronic phase as melatonin can diminish the inflammatory foci and cardiomyocyte damage in chronically infected mice (101). Despite the positive results in cardiac tissue, a recent study shows no diminishment in parasite replication and an accelerated parasitic release during the acute phase of infection in infected mice treated with melatonin (102). Therefore, more study is necessary to assess the risk-benefit use of melatonin in T. cruzi infection.
4. MELATONIN AND LEISHMANIA
Considered a neglected tropical disease, Leishmaniasis is caused by protozoan parasites from Leishmania (L.) genus (103, 104). Worldwide, Leishmaniasis causes approximately 20,000–30,000 deaths annually, and it is estimated that there are 12 million people infected (105). The parasite is transmitted by phlebotomine sandflies and is an obligatory-intracellular parasite, with part of the cycle in macrophages phagolysosomes (106, 107).
Laranjeira-Silva et al. (108) reported that the time of infection could alter the outcome of this infection by L. amazonensis in rats. The animals inoculated with the parasite in the dark phase had smaller lesions than the animals inoculated in the light phase. To confirm whether endogenous melatonin could interfere with the infectivity of L. amazonensis, a pre-treatment with luzindole (melatonin receptor antagonist) before melatonin treatment was performed, and they observed reduced lesions caused by the parasites. An in vitro invasion assay also observed a reduction in L. amazonensis infectivity in melatonin-treated-murine macrophages.
Emahallawy (109) observed that melatonin treatment decreases L. infantum growth in vitro. They also found that melatonin triggered the opening of the permeability transition pore (PTP), a structure present in mitochondria whose opening is related to cell death by compromising the mitochondrial membrane potential. Along with action in PTP, melatonin also impaired mitochondrial complex chains I, II, and III, components of the electron transport chain of L. infantum (109, 110). These actions of melatonin might be responsible for the decrease of parasite growth under melatonin treatment.
5. MELATONIN AND OTHER PARASITES
Toxoplasma gondii is the parasite responsible for toxoplasmosis disease. By treating T. gondii-infected rats with melatonin, Baltaci et al. (111) observed an increased leukocyte count in the bloodstream. In addition, T. gondii-infected macrophages treated with melatonin reduced parasite proliferation and apoptosis-induced death (112). In the absence of indoleamine-pyrrole-2,3-dioxygenase-1 (IDO1), the enzyme that catabolizes tryptophan, the T. gondii increases the biosynthesis of melatonin, which, interestingly, causes an increase in parasite growth and decreases levels of reactive oxygen species (ROS) (113). Further studies are necessary to better understand the action of internal and external melatonin in T. gondii.
Schistosomiasis is a disease caused by the parasite Schistosoma sp. The main complication of the disease is the formation of granuloma in the liver, causing liver fibrosis and portal hypertension (114). Knowing that the oxidative process is essential for the progression of schistosomiasis (114–117), El-Sokkari et al. (117) treated S. mansoni-infected rats with melatonin. The first parameter analyzed was the death rate of the infected mice, with a substantial reduction in the melatonin group. A decrease in granuloma formation was observed at the tissue level and diminished or prevented pathological changes caused by S. mansoni in the kidney, liver, and spleen. Melatonin reduced NO, inducible nitric oxide synthetase, and lipoperoxidation levels in S. mansoni-infected mice (117). The elevated level of ROS, including nitric oxide (NO), reflect the increased oxidation and tissue damage (118).
Opisthorchis viverrini is an inductive-cholangiocarcinoma parasite that is endemic in Southeast Asia (119, 120). Laothong et al. (121) observed that hamsters infected with O. viverrini treated with melatonin decreased liver injury, DNA damage, and increased nuclear factor-erythroid 2-related factor-2 (Nrf2) expression. Nrf2 is a transcription factor responsible for regulating response against oxidative damage (121). Also, melatonin caused the downregulation of oxidant genes, such as NF-ⲕB, iNOS, COX-2, and reduction of proinflammatory cytokine expression in parasitized animals (122).
The amoebiasis is due to the Entamoeba histolytica parasite. The infection leads to an inflammatory process with immune cell recruitment, triggering tissue damage (123). França-Botelho et al. (124) treated E. histolytica-infected hamsters with melatonin and observed decreased hepatic necrosis and inflammatory infiltrate while the increased activity of leukophagocytosis. These results indicate immunomodulatory effects of melatonin against infection caused by E. histolytica.
6. CONCLUSION
Melatonin, a host hormone, is capable of modulating parasitic infection by preventing tissue damage, regulating gene expression and inflammatory process, and acting as a free radical scavenger. So far, the influence of melatonin-related compounds as precursors, metabolites, and indolic molecules was studied in Plasmodium sp. parasites with interesting results, such as an increase in parasitemia and activation of intracellular signaling cascade. Therefore, further study of the net of intracellular players involved in this cascade is essential to bring light to the host-parasite relationship and is a promising path for further studies with other parasites. An understanding of the influence of melatonin and melatonin-related compounds in parasitic infections can help cast more light on host-parasite interactions and find a way to block parasite multiplication within the host.
ACKNOWLEDGEMENTS
We are grateful for the support of Fundação de Amparo à pesquisa do Estado de São Paulo (2017/08684-7). This study was also supported by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – Brasil (CAPES). RIH is CAPES fellow - Finance Code 88887.614839/2021-00. BKMD was a CNPq fellow Finance Code 142188/2017-4.
AUTHORSHIP
CRGS and JMP contributed to the critical review and approval of the text. RIH contributed to reading the literature, preparing the manuscript and figures. BKMD contributed by structuring the manuscript and revising it along with the figure.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
REFERENCES
Tan DX, Zheng X, Kong J, Manchester LC, Hardel R, Kim SJ, Xu X, Reiter RJ (2014) Fundamental issues related to the origin of melatonin and melatonin isomers during evolution: relation to their biological functions. Int. J. Mol. Sci. 15: 15858–15890. DOI: 10.3390/ijms150915858.
Galano A, Tan DX, Reiter RJ (2011) Melatonin as a natural ally against oxidative stress: a physicochemical examination. J. Pineal Res. 1: 1-16. DOI: 10.1111/j.1600-079X.2011.00916.x. PMID: 21752095.
Hardeland R, Poeggeler B (2008) Melatonin Beyond Its Classical Functions. Open Physiol. J. 1: 1-22. DOI: 10.2174/1874360900901010001.
Leite AA, Reiter RJ, Brandão JCM, Sakae TM, Marinho M, Camargo CR, Oliveira-Junior IS (2021) Melatonin can be, more effective than N-acetylcysteine, protecting acute lung injury induced by intestinal ischemia-reperfusion in rat model. Clinics 76: e2513 DOI: 10.6061/CLINICS/2021/E2513.
Ren BC, Zhang W, Zhang W, Ma JX, Pei F, Li BY (2021) Melatonin attenuates aortic oxidative stress injury and apoptosis in STZ-diabetes rats by Notch1/Hes1 pathway. J. Steroid Biochem. Mo.l Biol. 212: 105948. DOI: 10.1016/J.JSBMB.2021.105948.
Sadoughi F, Dana PM, Asemi Z, Shafabakhash R, Mohammadi S, Heidar Z, Mirzamoradi M, Targhazeh N, Mirzaei H (2022) Molecular and cellular mechanisms of melatonin in breast cancer. Biochimie S0300-9084(22)00067-0. DOI: 10.1016/j.biochi.2022.03.005.
Guerra J, Devesa J. (2022) Usefulness of Melatonin and Other Compounds as Antioxidants and Epidrugs in the Treatment of Head and Neck Cancer. Antioxidants 11 (1): 35. DOI: 10.3390/antiox11010035.
Ashrafizadeh M, Ahmadi Z, Yaribeygi H, Sathyapalan T, Jamialahmadi T, Sahebkar A (2021) Antitumor and Protective Effects of Melatonin: The Potential Roles of MicroRNAs. Adv. Exp. Med. Bio. 1328: 463-471. DOI: 10.1007/978-3-030-73234-9_3.1
Lim S, Park S, Koyanagi A, Yang JW, Jacob L, Yon DK, Lee SW, Kim MS, Il Shin J, Smith L (2022) Effects of exogenous melatonin supplementation on health outcomes: An umbrella review of meta-analyses based on randomized controlled trials. Pharmacol. Res. 176: 106052. DOI: 10.1016/j.phrs.2021.106052.
Reiter RJ, Sharma R, Simko F, Dominguez-Rodriguez A, Tesarik J, Neel RL, Slominski AT, Kleszczynski K, Martin-Gimenez VM, Manucha W, Cardinali DP (2022) Melatonin: highlighting its use as a potential treatment for SARS-CoV-2 infection. Cell. Mol. Life Sci. 3: 143. DOI: 10.1007/s00018-021-04102-3.
Tan DX, Reiter RJ (2022) Mechanisms and clinical evidence to support melatonin's use in severe COVID-19 patients to lower mortality. Life Sci. 294: 120368. DOI: 10.1016/j.lfs.2022.120368.
Li L, Gang X, Wang J, Gong X (2022) Role of melatonin in respiratory diseases. Exp. Ther. Med. 4: 271. DOI: 10.3892/etm.2022.11197.
Reppert SM, Weaver DR, Ebisawa T (1994) Cloning and characterization of a mammalian melatonin receptor that mediates reproductive and circadian responses. Neuron 13: 1177–1185. DOI: 10.1016/0896-6273(94)90055-8.
Reppert SM, Godson C, Mahle CD, Weaver DR, Slaugenhaupt SA, Gusella JF (1995) Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor. Proc. Natl. Acad. Sci. USA 92: 8734–8738. DOI: 10.1073/PNAS.92.19.8734.
Petit L, Lacroix I, de Coppet P, Strosberg AD, Jockers R (1999) Differential signaling of human Mel1a and Mel1b melatonin receptors through the cyclic guanosine 3’-5’-monophosphate pathway. Biochem. Pharmacol. 58: 633–639. DOI: 10.1016/S0006-2952(99)00134-3.
Lerner AB, Case JD, Takahashi Y, Lee TH, Mori W (1958) Isolation of melatonin, the pineal gland factor that lightens melanocytes. J. Am. Chem. Soc. 80: 2587. DOI: 10.1021/JA01543A060.
Reiter RJ, Tan DX, Kim SJ, Cruz MH (2014) Delivery of pineal melatonin to the brain and SCN: role of canaliculi, cerebrospinal fluid, tanycytes and Virchow-Robin perivascular spaces. Brain Struct.Funct. 6: 1873-87. DOI: 10.1007/s00429-014-0719-7.
Tan DX, Manchester LC, Reiter RJ (2016) CSF generation by pineal gland results in a robust melatonin circadian rhythm in the third ventricle as an unique light/dark signal. Med. Hypotheses. 86: 3-9. DOI: 10.1016/j.mehy.2015.11.018.
Bitar RD, Torres-Garza JL, Reiter RJ, Phillips WT (2021) Neural glymphatic system: Clinical implications and potential importance of melatonin. Mel. Res. 4: 551-565. DOI:https://doi.org/https://doi.org/10.32794/mr112500111.
Stefulj J, Hörtner M, Ghosh M, Schauenstein K, Rinner I, Wölfler A, Semmler J, Liebmann PM (2001) Gene expression of the key enzymes of melatonin synthesis in extrapineal tissues of the rat. J. Pineal Res. 4: 243-7. DOI: 10.1034/j.1600-079x.2001.300408.x.
Sanchez-Hidalgo M, de la Lastra CA, Carrascosa-Salmoral MP, Naranjo MC, Gomez-Corvera A, Caballero B, Guerrero JM (2009) Age-related changes in melatonin synthesis in rat extrapineal tissues. Exp. Gerontol. 5: 328-34. DOI: 10.1016/j.exger.2009.02.002.
Bubenik GA (2002) Gastrointestinal melatonin: localization, function, and clinical relevance. Dig. Dis. Sci. 10: 2336-48. DOI: 10.1023/a:1020107915919. PMID: 12395907.
Acuña-Castroviejo D, Escames G, Venegas C, Díaz-Casado ME, Lima-Cabello E, López LC, Rosales-Corral S, Tan DX, Reiter RJ (2014) Extrapineal melatonin: sources, regulation, and potential functions. Cell. Mol. Life Sci. 16: 2997-3025. DOI: 10.1007/s00018-014-1579-2.
Tan DX, Manchester LC, Liu X, Rosales-Corral SA, Acuna-Castroviejo D, Reiter RJ (2013) Mitochondria and chloroplasts as the original sites of melatonin synthesis: a hypothesis related to melatonin's primary function and evolution in eukaryotes. J. Pineal Res. 2: 127-38. DOI: 10.1111/jpi.12026.
Anderson G (2019) Gut Dysbiosis Dysregulates Central and Systemic Homeostasis via Decreased Melatonin and Suboptimal Mitochondria Functioning: Pathoetiological and Pathophysiological Implications. Mel. Res. 2: 70-85. DOI: 10.32794/mr11250022.
Anderson G (2020) Tumour Microenvironment: Roles of the Aryl Hydrocarbon Receptor, O-GlcNAcylation,Acetyl-CoA and Melatonergic Pathway in Regulating Dynamic Metabolic Interactions across Cell Types-Tumour Microenvironment and Metabolism. Int. J. Mol. Sci. 1: 141. DOI: 10.3390/ijms22010141. PMID: 33375613; PMCID: PMC7795031.
Klein DC (2007) Arylalkylamine N-acetyltransferase: “The timezyme.” J. Biol. Chem. 282: 4233–4237. DOI: 10.1074/jbc.R600036200.
Ganguly S, Coon SL, Klein DC (2002) Control of melatonin synthesis in the mammalian pineal gland: The critical role of serotonin acetylation. Cell Tissue Res. 309: 127–137. DOI: 10.1007/s00441-002-0579-y.
Ho AK, Thomas TP, Chik CL, Anderson WB, Klein DC (1988) Protein kinase C: Subcellular redistribution by increased Ca2+ influx. Evidence that Ca2+-dependent subcellular redistribution of protein kinase C is involved in potentiation of β-adrenergic stimulation of pineal cAMP and cGMP by K+ and A23187. J. Biol. Chem. 263: 9292–9297. DOI: 10.1016/s0021-9258(19)76538-7.
Sugden D, Vanecek J, Klein DC, Thomas TP, Anderson WB (1985) Activation of protein kinase C potentiates isoprenaline-induced cyclic AMP accumulation in rat pinealocytes. Nature 314: 359–361. DOI: 10.1038/314359a0.
Sugden LA, Sugden D, Klein DC (1987) Alpha 1-adrenoceptor activation elevates cytosolic calcium in rat pinealocytes by increasing net influx. J. Biol. Chem. 262: 741–745. DOI: 10.1016/S0021-9258(19)75847-5.
Ho AK, Klein DC (1987) Activation of alpha 1-adrenoceptors, protein kinase C, or treatment with intracellular free Ca2+ elevating agents increases pineal phospholipase A2 activity. Evidence that protein kinase C may participate in Ca2+-dependent alpha 1-adrenergic stimulation of pineal phospholipase A2 activity. J. Biol. Chem. 262: 11764–11770. DOI: 10.1016/S0021-9258(18)60877-4.
Sugden AL, Sugden D, Klein DC (1986) Essential role of calcium influx in the adrenergic regulation of cAMP and cGMP in rat pinealocytes. J. Biol. Chem. 261: 11608–11612. DOI: 10.1016/s0021-9258(18)67286-2.
Ganguly S, Gastel JA, Weller JL, Schwartz C, Jaffe H, Namboodiri MAA, Coon SL, Hickman AB, Rollag M, Obsil T, Beauverger P, Ferry G, Boutin JA, Klein DC (2001) Role of a pineal cAMP-operated arylalkylamine N-acetyltransferase14-3-3-binding switch in melatonin synthesis. Proc. Natl. Acad. Sci. USA 98: 8083-8088. DOI: 10.1073/PNAS.141118798.
Ganguly S, Weller JL, Ho A, Chemineau P, Malpaux B, Klein DC (2005) Melatonin synthesis: 14-3-3-dependent activation and inhibition of arylalkylamine N-acetyltransferase mediated by phosphoserine-205. Proc. Natl. Acad. Sci. USA 102: 1222-1227. DOI: 10.1073/PNAS.0406871102.
Pozdeyev N, Taylor C, Haque R, Chaurasia SS, Visser A, Thazyeen A, Du Y, Fu H, Weller J, Klein DC, Iuvone PM (2006) Photic regulation of arylalkylamine N-acetyltransferase binding to 14-3-3 proteins in retinal photoreceptor cells. J. Neurosci. 26: 9153–9161. DOI: 10.1523/JNEUROSCI.1384-06.2006.
World Health Organization (2020) World Malaria Report: 20 years of global progress and challenges. World Health Organization WHO/HTM/GM:299.
Sabbatani S, Fiorino S, Manfredi R (2010) The emerging of the fifth malaria parasite (Plasmodium knowlesi): a public health concern? Braz. J. Infect. Dis. 14: 299–309. DOI: 10.1590/s1413-86702010000300019.
Klein EY (2013) Antimalarial drug resistance: A review of the biology and strategies to delay emergence and spread. Int. J. Antimicrob. Agents 41: 311–317. DOI: 10.1016/j.ijantimicag.2012.12.007.
Mitchell GH, Bannister LH, Sinden RE (1988) Malaria parasite invasion: Interactions with the red cell membrane. Critic. Rev. Oncol. Hematol. 8: 255–310. DOI: 10.1016/S1040-8428(88)80011-8.
Cowman AF, Healer J, Marapana D, Marsh K (2016) Malaria: Biology and Disease. Cell 167: 610–624. DOI: 10.1016/J.CELL.2016.07.055.
Rijo-Ferreira F, Acosta-Rodriguez VA, Abel JH, Kornblum I, Bento I, Kilaru G, Klerman EB, Mota MM, Takahashi JS (2020) The malaria parasite has an intrinsic clock. Science 368: 746–753. DOI: 10.1126/science.aba2658.
Smith LM, Motta FC, Chopra G, Moch JK, Nerem RR, Cummins B, Roche KE, Kelliher CM, Leman AR, Harer J, Gedeon T, Waters NC, Haase SB (2020) An intrinsic oscillator drives the blood stage cycle of the malaria parasite Plasmodium falciparum. Science 368: 754–759. DOI: 10.1126/science.aba4357.
Trager W, Jensen JB (1976) Human malaria parasites in continuous culture. Science (New York, NY) 193: 673–675. DOI: 10.1126/SCIENCE.781840.
Hotta CT, Gazarini ML, Beraldo FH, Varotti FP, Lopes C, Markus RP, Pozzan T, Garcia CRS (2000) Calcium-dependent modulation by melatonin of the circadian rhythm in malarial parasites. Nat. Cell Biol. 2: 466-468. DOI: 10.1038/35017112.
Bagnaresi P, Alves E, da Silva HB, Epiphanio S, Mota MM, Garcia CRS (2009) Unlike the synchronous Plasmodium falciparum and P. chabaudi infection, the P. berghei and P. yoelii asynchronous infections are not affected by melatonin. Int. J. Gen. Med. 2: 47–55. DOI: 10.2147/ijgm.s3699.
Boyd GH (1929) Induced variations in the asexual cycle of Plasmodium cathemerium. Am. J. Epidemiol. 9: 81-187. DOI: 10.1093/oxfordjournals.aje.a121636.
David PH, Hommel M, Benichout J-C, Eisen HA, Silva LH (1978) Isolation of malaria merozoites: Release of Plasmodium chabaudi merozoites from schizonts bound to immobilized concanavalin A (rodent malaria/concanavalin A-Sepharose/merozoite isolation Proc. Natl. Acad. Sci. USA 75: 5081-5084. DOI: 10.1073/pnas.75.10.5081.
Alves E, Bartlett PJ, Garcia CRS, Thomas AP (2011) Melatonin and IP3-induced Ca2+ release from intracellular stores in the malaria parasite Plasmodium falciparum within infected red blood cells. J. Biol. Chem. 286: 5905–5912. DOI: 10.1074/jbc.M110.188474.
Beraldo FH, Garcia CRS (2005) Products of tryptophan catabolism induce Ca2+ release and modulate the cell cycle of Plasmodium falciparum malaria parasites. J. Pineal Res. 39: 224–230. DOI: 10.1111/j.1600-079X.2005.00249.x.
Schuck DC, Ribeiro RY, Nery AA, Ulrich H, Garcia CRS (2011) Flow cytometry as a tool for analyzing changes in Plasmodium falciparum cell cycle following treatment with indol compounds. Cytometry Part A 79 A: 959–964. DOI: 10.1002/cyto.a.21136.
Budu A, Peres R, Bueno VB, Catalani LH, Garcia CRS (2007) N1-acetyl-N2-formyl-5-methoxykynuramine modulates the cell cycle of malaria parasites. J. Pineal Res. 42: 261–266. DOI: 10.1111/j.1600-079X.2006.00414.x.
Schuck DC, Jordão AK, Nakabashi M, Cunha AC, Ferreira VF, Garcia CRS (2014) Synthetic indole and melatonin derivatives exhibit antimalarial activity on the cell cycle of the human malaria parasite Plasmodium falciparum. Eur. J. Med. Chem. 78: 375–382. DOI: 10.1016/J.EJMECH.2014.03.055
Dias BKM, Nakabashi M, Alves MRR, Portella DP, Santos BM, Almeida FCS, Ribeiro RY, Schuck DC, Jordão AK, Garcia CRS (2020) The Plasmodium falciparum eIK1 kinase (PfeIK1) is central for melatonin synchronization in the human malaria parasite. Melatotosil blocks melatonin action on parasite cell cycle. J. Pineal Res. 69: e12685. DOI: 10.1111/JPI.12685.
Beraldo FH, Almeida FM, da Silva AM, Garcia CR (2005) Cyclic AMP and calcium interplay as second messengers in melatonin-dependent regulation of Plasmodium falciparum cell cycle. J. Cell Biol. 170: 551–557. DOI: 10.1083/JCB.200505117.
Alves E, Bartlett PJ, Garcia CR, Thomas AP (2011) Melatonin and IP3-induced Ca2+ release from intracellular stores in the malaria parasite Plasmodium falciparum within infected red blood cells. J. Biol. Chem. 286: 5905–5912. DOI: 10.1074/JBC.M110.188474.
Furuyama W, Enomoto M, Mossaad E, Kawai S, Mikoshiba K, Kawazu SI (2014) An interplay between 2 signaling pathways: Melatonin-cAMP and IP 3-Ca2+ signaling pathways control intraerythrocytic development of the malaria parasite Plasmodium falciparum. Biochem. Biophys. Res. Commun. 446: 125–131. DOI: 10.1016/j.bbrc.2014.02.070.
Pecenin MF, Borges-Pereira L, Levano-Garcia J, Budu A, Alves E, Mikoshiba K, Thomas A, Garcia CRS (2018) Blocking IP3 signal transduction pathways inhibits melatonin-induced Ca2+ signals and impairs P. falciparum development and proliferation in erythrocytes. Cell Calcium 72: 81–90. DOI: 10.1016/j.ceca.2018.02.004.
Alves E, Nakaya H, Guimarães E, Garcia CRS (2021) Combining IP3 affinity chromatography and bioinformatics reveals a novel protein-IP3 binding site on Plasmodium falciparum MDR1 transporter. bioRxiv 2021.03.25.437059. DOI: 10.1101/2021.03.25.437059. Preprint, posted March 25, 2021.
Widmann C, Gibson S, Jarpe MB, Johnson GL (1999) Mitogen-activated protein kinase: Conservation of a three-kinase module from yeast to human. Physiol. Rev. 79: 143–180. DOI: 10.1152/PHYSREV.1999.79.1.143.
Garrington TP, Johnson GL (1999) Organization and regulation of mitogen-activated protein kinase signaling pathways. Curr. Opin. Cell Biol. 11: 211–218. DOI: 10.1016/S0955-0674(99)80028-3.
Raman M, Cobb MH (2003) MAP kinase modules: many roads home. Curr. Biol. 13: 886-888. DOI: 10.1016/J.CUB.2003.10.053.
Dorin D, Semblat JP, Poullet P, Alano P, Goldring JP, Whittle C, Patterson S, Chakrabarti D, Doerig C (2005) PfPK7, an atypical MEK-related protein kinase, reflects the absence of classical three-component MAPK pathways in the human malaria parasite Plasmodium falciparum. Mol. Microbiol. 55: 184–186. DOI: 10.1111/J.1365-2958.2004.04393.X.
Dorin-Semblat D, Sicard A, Doerig C, Ranford-Cartwright L, Doerig C (2008) Disruption of the PfPK7 gene impairs schizogony and sporogony in the human malaria parasite Plasmodium falciparum. Eukaryot. cell 7: 279–285. DOI: 10.1128/EC.00245-07.
Koyama FC, Ribeiro RY, Garcia JL, Azevedo MF, Chakrabarti D, Garcia CRS (2012) Ubiquitin Proteasome System and the atypical kinase PfPK7 are involved in melatonin signaling in Plasmodium falciparum. J. Pineal Res. 53: 147–153. DOI: 10.1111/J.1600-079X.2012.00981.X.
Bennink S, Pradel G (2019) The molecular machinery of translational control in malaria parasites. Mol. Microbiol. 112: 1658–1673. DOI: 10.1111/MMI.14388.
Lima WR, Moraes M, Alves E, Azevedo MF, Passos DO, Garcia CRS (2013) The PfNF-YB transcription factor is a downstream target of melatonin and cAMP signalling in the human malaria parasite Plasmodium falciparum. J Pineal Res 53: 145–153. DOI: 10.1111/j.1600-079X.2012.01021.x.
Maity SN, de Crombrugghe B (1998) Role of the CCAAT-binding protein CBF/NF-Y in transcription. Trends Biochem. Sci. 23: 174–178. DOI: 10.1016/S0968-0004(98)01201-8.
Kao CY, Tanimoto A, Arima N, Sasaguri Y, Padmanabhan R (1999) Transactivation of the human cdc2 promoter by adenovirus E1A. E1A induces the expression and assembly of a heteromeric complex consisting of the CCAAT box binding factor, CBF/NF-Y, and a 110-kDa DNA-binding protein. J. Biol. Chem. 274: 23043–23051. DOI: 10.1074/JBC.274.33.23043.
Hu Q, Bhattacharya C, Maity SN (2002) CCAAT binding factor (CBF) binding mediates cell cycle activation of topoisomerase IIalpha. Conventional CBF activation domains are not required. J. Biol. Chem. 277: 37191–37200. DOI: 10.1074/JBC.M205985200.
Lima WR, Holder AA, Garcia CRS (2013) Melatonin Signaling and Its Modulation of PfNF-YB Transcription Factor Expression in Plasmodium falciparum. Int. J. Mol. Sci. 14: 13704. DOI: 10.3390/IJMS140713704.
Lima WR, Tessarin-Almeida G, Rozanski A, Parreira KS, Moraes MS, Martins DC, Hashimoto RF, Galante PAF, Garcia CRS (2016) Signaling transcript profile of the asexual intraerythrocytic development cycle of Plasmodium falciparum induced by melatonin and cAMP. Genes Cancer 7: 323–339. DOI: 10.18632/genesandcancer.118.
Scarpelli PH, Tessarin‐Almeida G, Viçoso KL, Lima WR, Borges‐Pereira L, Meissner KA, Wrenger C, Rafaello A, Rizzuto R, Pozzan T, Garcia CRS (2019) Melatonin activates FIS1, DYN1, and DYN2 Plasmodium falciparum related‐genes for mitochondria fission: Mitoemerald‐GFP as a tool to visualize mitochondria structure. J. Pineal Res. 66: 14347–14352. DOI: 10.1111/JPI.12484.
Iwasawa R, Mahul-Mellier AL, Datler C, Pazarentzos E, Grimm S (2011) Fis1 and Bap31 bridge the mitochondria-ER interface to establish a platform for apoptosis induction. EMBO J. 30: 556–568. DOI: 10.1038/EMBOJ.2010.346.
Singh MK, Tessarin-Almeida G, Dias BKM, Pereira PS, Costa F, Przyborski JM, Garcia CRS (2021) A nuclear protein, PfMORC confers melatonin dependent synchrony of the human malaria parasite P. falciparum in the asexual stage. Sci. Rep. 11: 1–11. DOI: 10.1038/s41598-021-81235-2.
Moissiard G, Cokus SJ, Cary J, Feng S, Billi AC, Stroud H, Husmann D, Zhan Y, Lajoie BR, McCord RP, Hale CJ, Feng W, Michaels SD, Frand AR, Pellegrini M, Dekker J, Kim JK, Jacobsen SE (2012) MORC Family ATPases Required for Heterochromatin Condensation and Gene Silencing. Science 336: 1448–1451. DOI: 10.1126/science.1221472.
Weiser NE, Yang DX, Feng S, Kalinava N, Brown KC, Khanikar J, Freeberg MA, Snyder MJ, Csankovszki G, Chan RC, Gu SG, Montgomery TA, Jacobsen SE, Kim JK (2017) MORC-1 Integrates Nuclear RNAi and Transgenerational Chromatin Architecture to Promote Germline Immortality. Dev. Cell 41: 408-423.e7. DOI: 10.1016/j.devcel.2017.04.023.
Briquet S, Ourimi A, Pionneau C, Bernardes J, Carbone A, Chardonnet S, Vaquero C (2018) Identification of Plasmodium falciparum nuclear proteins by mass spectrometry and proposed protein annotation. PloS ONE 13: DOI: 10.1371/JOURNAL.PONE.0205596.
Pierce KL, Premont RT, Lefkowitz RJ (2002) Seven-transmembrane receptors. Nat. Rev. Mol. Cell Biol. 3: 639–650. DOI: 10.1038/NRM908.
Hall RA, Premont RT, Lefkowitz RJ (1999) Heptahelical receptor signaling: beyond the G protein paradigm. J. Cell Biol. 145: 927–932. DOI: 10.1083/JCB.145.5.927.
Madeira L, Galante PAF, Budu A, Azevedo MF, Malnic B, Garcia CRS (2008) Genome-wide detection of serpentine receptor-like proteins in malaria parasites. PLoS ONE 3: DOI: 10.1371/journal.pone.0001889.
Guha M, Maity P, Choubey V, Mitra K, Reiter RJ, Bandyopadhyay U (2007) Melatonin inhibits free radical-mediated mitochondrial-dependent hepatocyte apoptosis and liver damage induced during malarial infection. J. Pineal Res. 43: 372–381. DOI: 10.1111/j.1600-079X.2007.00488.x.
Ataide BJA, Kauffmann N, Mendes NSF, Torres MLM, Anjos LM. Passos ACF, Moraes SAS, Batista EJO, Herculano AM, Oliveira KRHM (2020) Melatonin prevents brain damage and neurocognitive impairment induced by plasmodium berghei anka infection in murine model of cerebral malaria. Front. Cell. Infect. Microbiol. 10: 541624 DOI: 10.3389/fcimb.2020.541624.
Kedarisetty CK, Samaga BL, Vidyasagar S, Venkataraman J (2020) Oral melatonin improves the detection of parasitemia in malaria. J. Infect. Dev. Ctries 14: 1327–1331. DOI: 10.3855/JIDC.12518.
Chagas disease - PAHO/WHO | Pan American Health Organization. https://www.paho.org/en/topics/chagas-disease. Accessed 17 Sep 2021.
Tyler KM, Engman DM (2001) The life cycle of Trypanosoma cruzi revisited. Int. J. Parasitol. 31: 472–481. DOI: 10.1016/s0020-7519(01)00153-9.
CDC - Chagas Disease. https://www.cdc.gov/parasites/chagas/index.html. Accessed 17 Sep 2021
Santos CD, Levy AM, Toldo MP, Azevedo AP, Prado JC (2007) Haematological and histopathological findings after ovariectomy in Trypanosoma cruzi infected mice. Vet. Parasitol. 143: 222–228. DOI: 10.1016/J.VETPAR.2006.08.038.
Santos CD, Caldeira JC, Toldo MPA, Prado JC (2005) Trypanosoma cruzi: Effects of repetitive stress during the development of experimental infection. Exp. Parasitol. 110: 96–101. DOI: 10.1016/J.EXPPARA.2005.03.009.
Santello FH, Frare EO, Santos CD, Caetano LC, Toldo MPA, Prado JC (2008) Suppressive action of melatonin on the TH-2 immune response in rats infected with Trypanosoma cruzi. J. Pineal Res. 45: 291–296. DOI: 10.1111/J.1600-079X.2008.00589.X.
Santello FH, Frare EO, dos Santos CD, Toldo MP, Kawasse LM, Zucoloto S, Prado JC (2007) Melatonin treatment reduces the severity of experimental Trypanosoma cruzi infection. J. Pineal Res. 42: 359–363. DOI: 10.1111/J.1600-079X.2007.00427.X.
Santello FH, Frare EO, Caetano LC, AlonsoToldo MP, Prado JC (2008) Melatonin enhances pro-inflammatory cytokine levels and protects against Chagas disease. J. Pineal Res. 45: 79–85. DOI: 10.1111/J.1600-079X.2008.00558.X.
Santello FH, del Vecchio Filipin M, Caetano LC, Brazão V, Caetano LN, Santos CD, AlonsoToldo MP, Prado JC (2009) Influence of melatonin therapy and orchiectomy on T cell subsets in male Wistar rats infected with Trypanosoma cruzi. J. Pineal Res. 47: 271–276. DOI: 10.1111/J.1600-079X.2009.00710.X.
Kuehn CC, Rodrigues Oliveira LG, Santos CD, Ferreira DS, Alonso Toldo MP, Albuquerque S, Prado JC (2009) Melatonin and dehydroepiandrosterone combination: does this treatment exert a synergistic effect during experimental Trypanosoma cruzi infection? J. Pineal Res. 47: 253–259. DOI: 10.1111/J.1600-079X.2009.00708.X.
Brazão V, Santello FH, Colato RP, Duarte A, Goulart A, Sampaio PA, Nardini V, Sorgi CA, Faccioli LH, Prado JC (2020) Melatonin down-regulates steroidal hormones, thymocyte apoptosis and inflammatory cytokines in middle-aged T. cruzi infected rats. Biochim. Biophys. Mol. Basis Dis 1866: 165914. DOI: 10.1016/J.BBADIS.2020.165914.
Brazão V, Colato RP, Santello FH, Vale GT, Gonzaga NA, Tirapelli CR, Prado JC (2018) Effects of melatonin on thymic and oxidative stress dysfunctions during Trypanosoma cruzi infection. J. Pineal Res. 65: e12510. DOI: 10.1111/JPI.12510.
Brazão V, Santello FH, Colato RP, Mazotti TT, Tazinafo LF, Toldo MPA, Vale GT, Tirapelli CR, Prado JC (2017) Melatonin: Antioxidant and modulatory properties in age-related changes during Trypanosoma cruzi infection. J. Pineal Res. 63: e12409. DOI: 10.1111/JPI.12409.
Brazão V, Filipin MDV, Santello FH, Caetano LC, Abrahão AMC, Toldo MPA, Prado JC (2011) Melatonin and zinc treatment: distinctive modulation of cytokine production in chronic experimental Trypanosoma cruzi infection. Cytokine 56: 627–632. DOI: 10.1016/J.CYTO.2011.08.037
Brazão V, Santello FH, Mdel FV, Azevedo AP, Toldo MP, Morais FR, Prado JC (2015) Immunoregulatory actions of melatonin and zinc during chronic Trypanosoma cruzi infection. J. Pineal Res. 58: 210–218. DOI: 10.1111/JPI.12207.
Oliveira LGR, Kuehn CC, dos Santos CD, Miranda MA, Costa CMB, Mendonça VJ, Prado JC (2013) Protective actions of melatonin against heart damage during chronic Chagas disease. Acta Trop. 128: 652–658. DOI: 10.1016/J.ACTATROPICA.2013.09.014.
Providello MV, Portapilla GB, Oliveira PAS, Silva CBP, Anchieta NF, Tirapelli CR, Albuquerque S (2020) Melatonin decreases circulating Trypanosoma cruzi load with no effect on tissue parasite replication. Can. J. Physiol. Pharmacol. 99: 795–802. DOI: 10.1139/CJPP-2020-0473.
Marsden PD (1986) Mucosal leishmaniasis (“espundia” Escomel, 1911). Trans. R. Soc. Trop. Med. Hyg. 80: 859–876. DOI: 10.1016/0035-9203(86)90243-9.
Ashford RW (2000) The leishmaniases as emerging and reemerging zoonoses. Int. J. Parasitol. 30: 1269–1281. DOI: 10.1016/S0020-7519(00)00136-3.
Alvar J, Vélez ID, Bern C, Herrero M, Desjeux P, Cano J, Jannin J, Boer M, Team the WHO Leishmaniasis Control (2012) Leishmaniasis worldwide and global estimates of its incidence. Plos ONE 7: e35671. DOI: 10.1371/JOURNAL.PONE.0035671.
Scott P, Novais FO (2016) Cutaneous leishmaniasis: immune responses in protection and pathogenesis. Nat. Rev. Immunol. 16: 581–592. DOI: 10.1038/nri.2016.72.
Gregory D, Oliver M (2005) Subversion of host cell signalling by the protozoan parasite Leishmania. Parasitology 130: S27–S35. DOI: 10.1017/S0031182005008139.
Laranjeira-Silva MF, Zampieri RA, Muxel SM, Floeter-Winter LM, Markus RP (2015) Melatonin attenuates Leishmania (L.) amazonensis infection by modulating arginine metabolism. J. Pineal Res. 59: 478–487. DOI: 10.1111/JPI.12279.
Elmahallawy EK, Jiménez-Aranda A, Martínez AS, Rodriguez-Granger J, Navarro-Alarcón M, Gutiérrez-Fernández J, Agil A (2014) Activity of melatonin against Leishmania infantum promastigotes by mitochondrial dependent pathway. Chem. Biol. Interact. 220: 84–93. DOI: 10.1016/J.CBI.2014.06.016.
Petronilli V, Penzo D, Scorrano L, Bernardi P, Di Lisa F (2001) The Mitochondrial Permeability Transition, Release of Cytochrome c and Cell Death: correlation with the duration of pore openings in situ. J. Biol. Chem. 276: 12030–12034. DOI: 10.1074/JBC.M010604200.
Baltaci AK, Bediz CS, Mogulkoc R, Kurtoglu E, Pekel A (2003) Effect of zinc and melatonin supplementation on cellular immunity in rats with toxoplasmosis. Biol. Trace Elem. Res. 96: 237–245. DOI: 10.1385/BTER:96:1-3:237.
Machado NI, dos Santos TAT, de Souza W, DaMatta RA, Seabra SH (2020) Treatment with melatonin induces a reduction of Toxoplasma gondii development in LLC-MK2 cells. Parasitol. Res. 119: 2703–2711. DOI: 10.1007/S00436-020-06766-5.
Majumdar T, Sharma S, Kumar M, Hussain MA, Chauhan N, Kalia I, Sahu AK, Rana VS, Bharti R, Haldar AK, Singh AP, Mazumder S (2019) Tryptophan-kynurenine pathway attenuates β-catenin-dependent pro-parasitic role of STING-TICAM2-IRF3-IDO1 signalosome in Toxoplasma gondii infection. Cell Death Dis. 10: (3): 161 DOI: 10.1038/S41419-019-1420-9.
Steinmann P, Keiser J, Bos R, et al (2006) Schistosomiasis and water resources development: systematic review, meta-analysis, and estimates of people at risk. Lancet Infect, Dis. 6: 411–425. DOI: 10.1016/S1473-3099(06)70521-7.
Abdallahi OM, Hanna S, de Reggi M, Gharib B (1999) Visualization of oxygen radical production in mouse liver in response to infection with Schistosoma mansoni. Liver 19: 495–500. DOI: 10.1111/J.1478-3231.1999.TB00082.X.
Caulfield JP, Lenzi HL, Elsas P, Dessein AJ (1985) Ultrastructure of the attack of eosinophils stimulated by blood mononuclear cell products on schistosomula of Schistosoma mansoni. Am. J. Pathol. 120: 380–390.
Kazura JW, Brito P de, Rabbege J, Aikawa M (1985) Role of granulocyte oxygen products in damage of Schistosoma mansoni eggs in vitro. J. Clin. Invest. 75: 1297–1307. DOI: 10.1172/JCI111830.
El-Sokkary GH, Omar HM, Hassanein AFMM, Cuzzocrea S, Reiter RJ (2002) Melatonin reduces oxidative damage and increases survival of mice infected with Schistosoma mansoni. Free Radic. Biol. Med. 32: 319–332. DOI: 10.1016/S0891-5849(01)00753-5.
Humans IWG on the E of CR to (1994) Infection with liver flukes (Opisthorchis viverrini, Opisthorchis felineus and Clonorchis sinensis). IARC Monogr. Eval. Carcinog. Risks Hum. 61: 121–175.
Sripa B, Kaewkes S, Sithithaworn P, Mairiang E, Laha T, Smout M, Pairojkul C, Bhudhisawasdi V, Tesana S, Thinkamrop B, Bethony JM, Loukas A, Brindley PJ (2007) Liver fluke induces cholangiocarcinoma. PLoS medicine 4: 1148–1155. DOI: 10.1371/JOURNAL.PMED.0040201.
Laothong U, Pinlaor P, Hiraku Y, Boonsiri P, Prakobwong S, Khoontawad J, Pinlaor S (2010) Protective effect of melatonin against Opisthorchis viverrini-induced oxidative and nitrosative DNA damage and liver injury in hamsters. J. Pineal Res. 49: 271–282. DOI: 10.1111/J.1600-079X.2010.00792.X.
Kim J, Cha YN, Surh YJ (2010) A protective role of nuclear factor-erythroid 2-related factor-2 (Nrf2) in inflammatory disorders. Mutat. Res. 690: 12–23. DOI: 10.1016/J.MRFMMM.2009.09.007.
Olivos-García A, Nequiz-Avendaño M, Tello E, Martínez RD, González-Canto A, López-Vancell R, García de León MC, Montfort I, Pérez-Tamayo R (2004) Inflammation, complement, ischemia and amoebic survival in acute experimental amoebic liver abscesses in hamsters. Exp. Mol. Pathol. 77: 66–71. DOI: 10.1016/J.YEXMP.2003.09.003.
França-Botelho AC, França JL, Oliveira FM, Franca EL, Honário-França AC, Caliari MV, Gomes MA (2011) Melatonin reduces the severity of experimental amoebiasis. Parasit. Vectors 4: 62. DOI: 10.1186/1756-3305-4-62.
This work is licensed under a Creative Commons Attribution 4.0 International License