Next Article in Journal
Kinetoplast Genome of Leishmania spp. Is under Strong Purifying Selection
Next Article in Special Issue
Parasite Detection in Visceral Leishmaniasis Samples by Dye-Based qPCR Using New Gene Targets of Leishmania infantum and Crithidia
Previous Article in Journal
Cell Aggregation Capability of Clinical Isolates from Candida auris and Candida haemulonii Species Complex
Previous Article in Special Issue
In Vitro Drug Susceptibility of a Leishmania (Leishmania) infantum Isolate from a Visceral Leishmaniasis Pediatric Patient after Multiple Relapses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Schistosoma and Leishmania: An Untold Story of Coinfection

by
Genil Mororó Araújo Camelo
,
Jeferson Kelvin Alves de Oliveira Silva
,
Stefan Michael Geiger
,
Maria Norma Melo
and
Deborah Aparecida Negrão-Corrêa
*
Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
*
Author to whom correspondence should be addressed.
Trop. Med. Infect. Dis. 2023, 8(8), 383; https://doi.org/10.3390/tropicalmed8080383
Submission received: 29 June 2023 / Revised: 17 July 2023 / Accepted: 20 July 2023 / Published: 27 July 2023
(This article belongs to the Special Issue Emerging Topics in Leishmaniasis Research)

Abstract

:
A remarkable characteristic of infectious diseases classified as Neglected Tropical Diseases (NTDs) is the fact that they are mostly transmitted in tropical and subtropical regions with poor conditions of sanitation and low access to healthcare, which makes transmission areas more likely to overlap. Two of the most important NTDs, schistosomiasis and leishmaniasis, despite being caused by very different etiological agents, have their pathogenesis heavily associated with immune-mediated mechanisms, and Schistosoma spp. and Leishmania spp. have been shown to simultaneously infect humans. Still, the consequences of Schistosoma–Leishmania coinfections remain underexplored. As the inflammatory processes elicited by each one of these parasites can influence the other, several changes have been observed due to this coinfection in naturally infected humans, experimental models, and in vitro cell assays, including modifications in susceptibility to infection, pathogenesis, prognostic, and response to treatment. Herein, we review the current knowledge in Schistosoma–Leishmania coinfections in both human populations and experimental models, with special regard to how schistosomiasis affects tegumentary leishmaniasis, discuss future perspectives, and suggest a few steps to further improve our understanding in this model of parasite–host–parasite interaction.

1. Often Forgotten, Never Gone: Neglected Tropical Diseases

People turn a blind eye daily, and it is no different when it comes to public health issues. Frequently forgotten and away from the spotlight, Neglected Tropical Diseases (NTDs) impose a devastating burden on the most vulnerable, marginalized populations [1]. They are most prevalent in tropical areas and affect over 1.7 billion people every year, who are exposed to pathogens, as well as their vectors and reservoirs [2]. Several targets were set by the World Health Organization (WHO) to prevent, control, eliminate or eradicate NTDs worldwide in 2020 [3], but some were not achieved, leaving us now with both new and old challenges heading towards 2030 [1]. In addition, the COVID-19 pandemic posed an unexpected, complicating factor in controlling NTDs. Health authorities have shifted their attention towards COVID-19, which lead to severe disruptions in NTD activities, including the diversion of financial resources, reassignment of personnel, discontinuation of monitoring programs, delays in diagnosis and treatment, and the overall collapse of health systems [2,4]. Among the control programs impacted by the pandemic, the ones aimed at controlling schistosomiasis and leishmaniasis were potentially two of the most severely hindered [5,6,7,8].
Schistosomiasis, a parasitic disease due to the infection by the trematodes from the genus Schistosoma, is endemic to 78 countries and caused a burden of 2.5 million disability-adjusted life years (DALYs) in 2016, leaving roughly 241.3 million people in need of mass drug administration in 2020 [2,9]. This genus comprises several species that can infect humans. S. mansoni is the only one present in the Americas; S. japonicum and S. mekongi affect parts of East and Southeast Asia; and Western Asia and Africa are afflicted by S. mansoni, S. haematobium, and S. intercalatum, with Sub-Saharan Africa being the most burdened [10].
Several clinical manifestations are attributed to schistosomiasis and, as an immunopathology, most of its symptoms are a side effect of the host’s own immune response. Although the granulomatous reaction is of utmost importance to contain toxic antigens secreted by trapped schistosome eggs, they are also the main cause of chronic pathology in this infection [11,12]. S. mansoni, S. intercalatum, S. mekongi, and S. japonicum infections mainly affect the intestines, liver, and spleen, while schistosomiasis caused by S. haematobium mostly compromises the urogenital system [13,14].
Leishmaniasis, in turn, is a group of diseases caused by protozoans from the genus Leishmania, divided into skin and systemic manifestations. Species such as L. braziliensis, L. amazonensis, L. major, and L. tropica mostly cause tegumentary leishmaniasis, including cutaneous and mucosal manifestations of leishmaniasis, while L. infantum and L. donovani are causative agents of visceral leishmaniasis [15,16,17,18,19,20,21]. Additionally, cases of post-kala-azar dermal leishmaniasis, a cutaneous manifestation, are reported after visceral leishmaniasis by L. donovani [15,16].
Tegumentary leishmaniasis is endemic to 89 countries and exhibited 221,953 cases in 2021, 99% of which happened in the Eastern Mediterranean, Argelia, and the Americas together [1,22,23]. However, its actual incidence has been estimated to be 2.5 to 4.3 times higher than what is reported [24]. Its clinical presentations affect the host’s integument, varying widely and being related to both the Leishmania spp. and the host’s immune response, ranging from self-healing localized ulcers to highly disfiguring mucosal lesions, due to extensive immune-driven tissue destruction, or diffuse nodular lesions, due to an anergic response [15,16,25].
Visceral leishmaniasis, on the other hand, is endemic to 80 countries and exhibited 11,743 cases in 2021, with three eco-epidemiological hotspots, East Africa, the Indian Subcontinent, and Brazil, reporting 94% of the cases that year [1,22,23]. In addition, although less prevalent than tegumentary leishmaniasis, this form is exceptionally lethal, leading to death in 95% of untreated symptomatic cases [1,24]. It mainly compromises internal organs such as the liver, spleen, and bone marrow, causing fever, anemia, hepatosplenomegaly, cachexia, and immunosuppression [16]. Pathogenesis has been related to parasite persistence, and an altered balance in different types of immune response [26,27].
As we resume activities aimed at controlling NTDs, we must bear in mind that the biological, geographical, and social contexts shared by several NTDs expose affected populations to a vast number of pathogens at the same time [1,2]. Therefore, it is reasonable to consider that a significant number of individuals in such areas are subjected to coinfections [28,29,30]. In addition, since the outcome of schistosomiasis and leishmaniasis is closely associated with the type and intensity of the host’s immune response during infection, it stands to reason that the immune response during coinfections may be altered to the extent that the prognosis of the diseases is modified [28,29,30]. Herein, we discuss the current knowledge on Schistosoma–Leishmania coinfections, an often-overlooked association, which may have profound consequences for the morbidity and treatment of either parasitic infection.

2. Two against the Host: The Influence of Schistosomiasis and Leishmaniasis on One Another

Coinfections with different species are a possibility worldwide as several human-infecting species of Schistosoma and Leishmania have distinct transmission areas. Coinfections with S. mansoni would be the only ones present in the Americas, while this and other species, e.g., S. haematobium, could be involved in coinfections in other regions, such as Africa and Western Asia [10]. Until now, only coinfections with S. mansoni and Leishmania spp. have been demonstrated in humans, mostly in Brazil [31,32,33,34] but also in Israel [35]. In this topic, we will explore what we know regarding the consequences of Schistosoma–Leishmania coinfections human populations.
The potential of schistosomiasis to interfere with other inflammatory diseases in humans, such as allergies, inflammatory bowel disease, toxoplasmosis, and malaria [36,37,38], has been investigated over the years, although not as extensively as in experimental models. However, much is left to uncover regarding coinfections with Leishmania spp.
Schistosomiasis seems to be detrimental to the treatment of tegumentary leishmaniasis. In the rural community of Corte de Pedra, Bahia, an endemic region in Northeastern Brazil, 88.3% of the people with active American tegumentary leishmaniasis (ATL) had helminth coinfections, with 16.7% being coinfected with S. mansoni [31]. The coinfected patients had an unsatisfactory response to antimonial therapy when compared with helminth-free individuals, with a higher frequency of persistent lesions, strongly associating helminth coinfections with a delayed lesion resolution [31]. A following randomized, double-blind, placebo-controlled trial in the same area found that early anthelminthic treatment did not improve on the subsequent antileishmanial therapy of L. braziliensis-coinfected patients [39]. In the State of Rio de Janeiro, Southeastern Brazil, 10% of the patients with American tegumentary leishmaniasis were infected with S. mansoni, and a higher frequency of therapeutic failure and relapse in helminth–Leishmania coinfected individuals was found as well [34]. Moreover, in a more recent case report, a coinfected patient from São Paulo, Southeastern Brazil, presented atypical manifestations of both schistosomiasis and ATL, and responded poorly to antimony treatment even after being treated with oxamniquine and praziquantel [33]. As indicated by O’Neal et al. [31], the helminth–ATL coinfected patients showed higher levels of serum immunoglobulin(Ig)E in comparison with ones with only ATL, indicating a higher interleukin(IL)-4 production in coinfected individuals that would contribute to these negative outcomes of hosts when faced with multiple infections.
The immune responses elicited by both parasites, summarized in Figure 1, are rather complex. During S. mansoni development in the vertebrate host, a predominant type 1 response is stimulated against cercariae penetration, schistosomula migration, and adult worms [40,41,42], followed mostly by a type 2 profile after oviposition begins [43,44,45,46,47]. In parallel, a regulatory response is developed, which takes over in the chronic phase of schistosomiasis [48,49,50,51]. In contrast, in tegumentary leishmaniasis, an exacerbated T helper(Th)1-polarized response is related to increased tissue destruction in mucosal leishmaniasis [52,53,54], while the lack of an appropriate type 1 response and the predominance of Th2 and regulatory mechanisms lead to unchecked amastigote proliferation in diffuse cutaneous leishmaniasis [55,56,57,58]. Therefore, it is reasonable to expect that coinfected individuals could ultimately present a different disease outcome prevalence due to modified immune-mediated processes [28].
In vitro studies have shed some light on how immune response is altered in different Schistosoma–Leishmania coinfection models. The stimulation of peripheral blood mononuclear cells (PBMC) from ATL patients with soluble Leishmania antigens (SLA) in the presence or absence of different adult S. mansoni antigens has yielded interesting results [59,60]. Although each of the worm antigens generated different effects, the costimulation overall induced the production of IL-10, while inhibiting interferon(IFN)-γ [59]. Moreover, the presence of worm antigens decreased the expression of human leukocyte antigen(HLA)-DR, CD80, and CD86 in monocytes, while increasing the expression of cytotoxic T-lymphocyte-associated protein(CTLA)-4 in CD4+ T lymphocytes, and the frequency of CD4+CD25highFoxP3+ T cells [60]. Together, these results indicate that being exposed to adult S. mansoni antigens may hamper antigen presentation in ATL patients.
Similarly, by costimulating monocyte-derived dendritic cells (MoDCs) harvested from ATL patients with Sm29, an adult S. mansoni antigen, and SLA, the frequency of regulatory markers increased, while the populations of CD40+, IL-12+, and tumor necrosis factor(TNF)+ remained similar [61]. When co-cultures of MoDCs with autologous lymphocytes from patients with schistosomiasis were infected in vitro with L. braziliensis, a lack of activation and induction of the regulatory profile were also observed both in MoDCs and in CD4+ and CD8+ cells, which made MoDCs more susceptible to the in vitro infection [62]. Conversely, the stimulation of MoDCs from ATL patients with recombinant Sm29 did not increase their susceptibility to L. braziliensis in the presence of autologous lymphocytes in spite of the similarly induced regulatory profile [63]. Thus, even though the S. mansoni infection itself seems to increase host-cell susceptibility to L. braziliensis, schistosome antigens could potentially be used to decrease inflammation and aid the healing process during ATL [63].
Despite our current knowledge regarding the immune response to and pathogenesis of schistosomiasis and leishmaniasis separately, and what we may expect to change in a coinfection context, the immunopathology during human Schistosoma–Leishmania coinfections remains largely underexplored. The number and size of typical cutaneous lesions seem not to be affected regardless of helminth coinfections [31], but a case report describes a coinfected individual with unusual clinical presentations, with abdominal skin papules containing S. mansoni eggs, while Leishmania sp. lesions manifested as a crusted, infiltrated erythematous plaque with irregular borders [33]. Therefore, research on atypical manifestations of schistosomiasis and leishmaniasis in coinfected patients may be promising. Regarding the most severe forms of tegumentary leishmaniasis, and helminth coinfections, including S. mansoni, in ATL patients from Rio de Janeiro were shown to increase the prevalence of mucosal leishmaniasis [34]. However, it remains to be seen whether schistosomiasis itself could affect the incidence of mucosal or diffuse leishmaniasis.
Compared with tegumentary leishmaniasis, how schistosomiasis influences visceral leishmaniasis has not been addressed nearly as much. As pathogenesis in visceral leishmaniasis involves chemokine expression and the migration of immune cells in the liver and spleen [26,27], an infection with another parasite such as S. mansoni, which also alters cytokine profiles and cellular infiltration in these organs [47], is expected to heavily affect this process. In this context, it is worth mentioning a case report of enteropathic visceral leishmaniasis with concomitant S. mansoni infection, with persistent diarrhea, the presence of L. infantum amastigotes in duodenal biopsies, and relapses [64]. In addition, a case report of an Eritrean refugee in Israel suggests that S. mansoni in coinfection with a species from the L. donovani complex may exacerbate spleen-related pathology [35], but the amount of data we currently have is too small for anyone to imply that there is any causal relation. Furthermore, the fact that a case of Schistosoma–Leishmania coinfection has been diagnosed in Israel, a country not endemic for schistosomiasis, exemplifies that schistosomiasis and leishmaniasis are not only a problem in endemic countries. Therefore, policymaking in disease control should always take into consideration current migration processes.
In addition, the increased in vitro susceptibility to Leishmania sp. due to S. mansoni coinfection or the exposure to worm antigens may jeopardize the control of leishmaniasis. In another coinfection context, visceral leishmaniasis patients coinfected with the human immunodeficiency virus (HIV) can present high parasite loads in the skin and a higher frequency of post-kala-azar dermal leishmaniasis [65,66]. Therefore, they could potentially act like superspreaders due to the increased likelihood of infecting sand flies in addition to serving as reservoirs to drug-resistant Leishmania spp. [23,66]. It is not certain whether coinfections with Schistosoma spp. could pose the same threat or not since we currently do not know how Schistosoma–Leishmania coinfections influence the presence of amastigotes in the human tissues accessible by sand flies; however, the existing reports of diminished therapeutic efficacy in Schistosoma–Leishmania coinfections are becoming particularly worrying [31,33,39,64]. Therefore, in vivo peripheral parasite load must be addressed in forthcoming studies.
As for the impact of leishmaniasis on schistosomiasis in humans, we still know very little. Since schistosomula have been shown to be controlled by type 1 immune responses in the vertebrate host [40,41,42,67], the typical Th1 response elicited by Leishmania spp. infections [15,25,52] could theoretically favor the killing of immature stages in a Schistosoma–Leishmania coinfection setting. However, a study conducted by our group in an endemic region with the active transmission of Leishmania sp. and S. mansoni in Minas Gerais, Southeastern Brazil, showed that the prevalence of schistosomiasis was higher in individuals who presented a history of ATL [32]. Regarding the serum immune response, this condition was positively associated with an IL-27 response, while negatively associated with IL-17 and CXCL10, suggesting that a Leishmania-driven modulation could affect the host’s susceptibility to S. mansoni [32]. However, the most important take-home message from this study may be that simultaneous coinfection is not a requirement, as changes in the immune system caused by previous ATL still affect posterior S. mansoni susceptibility. Interestingly, this agrees with the fact that early anthelminthic treatment does not seem to improve the outcome of ATL [39], since helminth infections can also induce long-lasting effects on the host’s immune response [68,69,70,71,72]. Considering that people who live in coendemic areas may eventually come across these pathogens at different times, the lingering immunological shifts caused by the first infection could reverberate in future diseases for a yet-undetermined period of their lives. Thus, not only coinfections deserve attention, but non-concomitant parasite–immune system–parasite interactions do as well.
Despite the valuable knowledge provided by studies in human populations, some caveats must be pointed out. Up until now, the studies concerning the topic have been either cross-sectional or started after the onset of both infections, which makes it nearly impossible to determine the time and sequence of infections. Furthermore, other coinfections present in the studied populations, such as other helminthiasis and protozoan infections, might obscure the effect that a single species of parasite has. Therefore, some immunopathological aspects may not be easily explored in human populations. With that in mind, these limitations can be overcome with the use of experimental models, which provide a more homogenous, controlled environment that can yield new insights into the impacts and mechanisms involved in the Schistosoma–Leishmania coinfection.

3. Making Heads and Tails: Experimental Coinfection Models

Indeed, some questions raised from human studies have been better investigated with in vivo and ex vivo studies using mice, a host susceptible to infection with several Leishmania spp. and S. mansoni [25,73,74]. Although not extensive in number, these studies have slowly but steadily contributed to what we know about Schistosoma–Leishmania coinfections. Most published studies were performed using L. major and S. mansoni, with only a few exceptions, and some data still show contrasting results.
The experimental coinfection with L. mexicana in outbred mice that were previously infected by S. mansoni (60 days before) resulted in a shorter incubation period, with lesions appearing earlier in coinfected mice than in L. mexicana-monoinfected ones [75]. Meanwhile, C57BL/6 mice coinfected with L. major after 2 weeks of infection with S. mansoni presented larger lesions, but with delayed growth and resolution, which coincided with an impaired ability to control the parasite, resulting in higher L. major parasite loads [76]. Similarly, the concomitant infection of BALB/c mice with S. mansoni and L. major resulted in larger footpad lesions after 10 weeks and reduced mortality [77].
Popliteal lymph node cells harvested from C57BL/6 mice coinfected with S. mansoni and L. major also showed a different immune response after in vitro stimulation with SLA [76]. The results of La Flamme et al. [76] show that levels of IFN-γ, TNF-α, and nitric oxide (NO) were lower at 4 weeks of coinfection (6 weeks post S. mansoni infection, 4 weeks post L. major infection), with increased levels of IL-4. However, this profile was reversed from the 8th week post-coinfection onwards. Moreover, peritoneal macrophages from 6–8-week-schistosome-infected mice were unable to exert leishmanicidal activity in vitro when stimulated with IFN-γ, with a defective production of NO compared with macrophages from non-infected mice.
In contrast, no impact on lesion development kinetics was observed when the L. major infection happened 8 weeks after the S. mansoni infection or at the same time, with BALB/c mice developing chronic lesions, while C57BL/6 mice evolved to spontaneous healing [78,79]. In this case, cytokine measurement in popliteal lymph node cell culture by RT-PCR revealed that coinfected BALB/c mice expressed less IL-4 and more IL-10, while in C57BL/6 mice, the IFN-γ expression remained unchanged regardless of the coinfection, which is expected based on the clinical manifestations presented by both mouse strains [78].
These contrasting results may be due to differences in infective loads, the time of coinfection, the mouse strain, and the parasite species and strain, which are summarized in Table 1. A decisive factor in experimental Schistosoma–Leishmania coinfections seems to be the time of infection, as L. major coinfections that happen within the first weeks of the schistosome infection [76,77] were shown to alter the development of tegumentary leishmaniasis, while infection after 8 weeks of S. mansoni did not [78]. However, despite happening at approximately the same time after S. mansoni infection, coinfection with L. major [78] or L. mexicana [75] presented different outcomes. This only reaffirms the need to study different Leishmania spp., since the lack of changes in the pathogenesis or immune response by analyzing a single species should not be regarded as truth for others. Thus, studies regarding Schistosoma sp. coinfections with L. braziliensis and L. amazonensis, two of the main etiological agents of ATL [18,52,80,81], and viscerotropic Leishmania spp. are of utmost importance.
As for visceral leishmaniasis, it was found that L. donovani can thrive in both the liver and spleen of mice coinfected with S. mansoni. C57BL/6 mice infected with L. donovani 8 weeks after S. mansoni infection showed increased L. donovani parasite burdens in these organs, with the multiplication of amastigotes in egg granulomas associated with the lower production of NO and IFN-γ, and higher levels of IL-4 and IL-10 [82]. Additionally, once again, the impact of leishmaniasis on experimental schistosomiasis has been given less attention in comparison with the other way around. No changes in adult-worm load or eggs retained in the liver were observed in coinfected C57BL/6 mice when L. major infection happened 2 weeks after S. mansoni infection [76].
The impact of Schistosoma–Leishmania coinfections on the treatment success of schistosomiasis and leishmaniasis has also been investigated in experimental models. The combined treatment of L. major-and-S. mansoni-coinfected BALB/c mice using Pentostam and praziquantel was more effective than monotherapy in reducing skin lesions, Leishman-Donovan Units, and adult-S. mansoni recovery [79]. It is important to point out that it is unclear if the contrasting results regarding combined therapy in humans [39] and experimental models [79] is a product of different Leishmania spp. infections (L. braziliensis vs. L. major), a different leishmanicidal drug of choice (Glucantime vs. Pentostam), other coinfections (coinfection with several parasites vs. only S. mansoni), host-related factors or another variable. Thus, it may not be reasonable to directly compare these results, making it clear that more studies are needed to determine the best course of action when it comes to treating coinfected patients.
However, it is important to mention that experimental models also have their own caveats. Although mice and their several strains can reflect both self-healing and chronic skin lesions in tegumentary leishmaniasis [25], they may not replicate faithfully atypical or more severe forms of this disease, such as diffuse or mucosal leishmaniasis. Alternatively, metastatic forms of tegumentary leishmaniasis may be more closely evaluated by using trauma-induced metastasis [83]. In addition, Syrian golden hamsters (Mesocricetus auratus) are highly susceptible to both dermotropic and viscerotropic Leishmania spp., manifesting lesions similar to those in humans [84,85,86]. Other non-murine models, namely dogs, even though useful for replicating mucocutaneous disease [87], may not be appropriate for experimental S. mansoni infections [88].
As for Schistosoma spp., S. mansoni can infect mice and golden hamsters [73,74,89]. Regarding other models of coinfection, golden hamsters have also been used for S. mansoniEntamoeba histolytica coinfection studies [90]. The limitations become more troubling when we consider that we still lack an accessible experimental model for S. haematobium infections, the etiologic agent of urogenital schistosomiasis. Rats, guinea pigs, and rabbits are not suitable hosts for this parasite, while mice and golden hamsters are only partially susceptible [73,74,91]. Although S. haematobium completes its life cycle in these two rodents, urinary involvement is absent in mice and inconsistent in hamsters [91]. Therefore, S. haematobium–Leishmania coinfections would be mostly limited to human studies and non-rodent models, which may further postpone advances in understanding changes in immunopathology during this coinfection and unraveling the involved mechanisms. This would not apply to S. japonicum and S. intercalatum, as they successfully infect mice [73].
A couple of unusual experimental models could be an alternative to work around some of the limitations presented by the more common rodent models. Some wild rodents, such as Nectomys spp. and Holochilus spp., can be maintained in laboratory and have been shown to be susceptible to Leishmania spp. and S. mansoni infections, and can also be found naturally infected [92,93,94,95,96]. Finally, while requiring a more sophisticated, expensive structure to maintain when compared with murine models, non-human primates might actually be a rather suitable model for the study of Schistosoma–Leishmania coinfections. First, several non-human primates are susceptible to both Leishmania spp. and Schistosoma spp. Additionally, L. braziliensis not only can induce chronic lesions in rhesus monkeys (Macaca mulatta) but they can also develop metastatic skin and mucosal lesions [97], providing a model for mucosal leishmaniasis under a Schistosoma-coinfection context. Diffuse leishmaniasis, as far as we know, has yet to be observed in non-human primates; however, it has been shown that M. mulatta is susceptible to infections with L. amazonensis, the main etiological agent of this clinical manifestation [18,98]. As for schistosomiasis, non-human primates, such as Cercopithecus spp., are susceptible to S. mansoni and S. haematobium infections both naturally and in a laboratory setting [99,100,101]. Moreover, the experimental infection of C. fuliginosus with S. haematobium more closely mimics human urogenital disease, with adult worms being present in prostatic, vesical, and uterine plexuses, with a high deposition of eggs in the bladder and uterus [101]. Therefore, these unusual models could be considered for studies on Schistosoma–Leishmania coinfections, especially when typical murine models are not appropriate.

4. Concluding Remarks and Future Directions

Even though it is not frequently given proper attention, Schistosoma–Leishmania coinfections could be more common than one would expect. Thus, this should be kept in mind when establishing control measures for both schistosomiasis and leishmaniasis. However, in the event that this coinfection becomes a reality when organizing control strategies for these diseases, much is still left to be uncovered.
We have yet to assess how widespread and numerous Schistosoma–Leishmania coinfections really are in different parts of the globe. Local studies that correlate the incidences of schistosomiasis and leishmaniasis can indicate areas where coinfections are more likely to occur, pointing research groups and authorities towards populations with a higher chance of presenting coinfected individuals. Longitudinal studies, on the other hand, provide an important tool in controlling the order of infection in endemic areas. In addition, the impact that this coinfection has on the transmission, burden, and treatment of such diseases must be better understood and would be a valuable aid in decision-making.
Studies using different Leishmania and Schistosoma species are essential, as, based on our current knowledge, interactions are likely different depending on the parasite species. Experimental coinfections could allow us to assess not only how the pathogenesis and parasite load change when different species are involved but also could provide a model which we can use to evaluate if Schistosoma–Leishmania coinfections can affect the transmission of these parasites, especially their infectivity to sand flies. Moreover, a better understanding of the immune mechanisms involved in this process could not only pave the way for more successful interventions in coinfected individuals but also potentially provide a better understanding of each infection separately.

Author Contributions

Conceptualization, G.M.A.C., J.K.A.d.O.S. and D.A.N.-C.; resources, D.A.N.-C.; writing—original draft preparation, G.M.A.C.; writing—review and editing, G.M.A.C., J.K.A.d.O.S., S.M.G., M.N.M. and D.A.N.-C.; visualization, G.M.A.C. and D.A.N.-C.; supervision, D.A.N.-C.; project administration, D.A.N.-C.; funding acquisition, D.A.N.-C. All authors have read and agreed to the published version of the manuscript.

Funding

Financial support for this work was partially provided by the Fundação de Amparo à Pesquisa do Estado de Minas Gerais (FAPEMIG-Brazil, grant #APQ 01637-17). D.A.N.-C. was supported by a fellowship from Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq/Brazil). G.M.A.C. was supported by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES/Brazil). The APC was partially funded by Pró-Reitoria de Pesquisa, Universidade Federal de Minas Gerais (PRPq/UFMG) and Programa de Pós-Graduação em Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (PPGPar/ICB/UFMG).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Acknowledgement is due to José Carlos dos Reis and Elizabete de Lacorte Marques for the technical support provided.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Ending the Neglect to Attain the Sustainable Development Goals: A Road Map for Neglected Tropical Diseases 2021–2030; World Health Organization: Geneva, Switzerland, 2020. [Google Scholar]
  2. WHO. Neglected tropical diseases: Impact of COVID-19 and WHO’s response—2021 update. Wkly. Epidemiol. Rec. 2021, 96, 461–468. [Google Scholar]
  3. WHO. Accelerating Work to Overcome the Global Impact of Neglected Tropical Diseases—A Roadmap for Implementation; World Health Organization: Geneva, Switzerland, 2012. [Google Scholar]
  4. Ung, L.; Stothard, J.R.; Phalkey, R.; Azman, A.S.; Chodosh, J.; Hanage, W.P.; Standley, C.J. Towards global control of parasitic diseases in the COVID-19 era: One Health and the future of multisectoral global health governance. Adv. Parasitol. 2021, 114, 1–26. [Google Scholar] [CrossRef] [PubMed]
  5. Brooker, S.J.; Ziumbe, K.; Negussu, N.; Crowley, S.; Hammami, M. Neglected tropical disease control in a world with COVID-19: An opportunity and a necessity for innovation. Trans. R. Soc. Trop. Med. Hyg. 2021, 115, 205–207. [Google Scholar] [CrossRef]
  6. Kura, K.; Ayabina, D.; Toor, J.; Hollingsworth, T.D.; Anderson, R.M. Disruptions to schistosomiasis programmes due to COVID-19: An analysis of potential impact and mitigation strategies. Trans. R. Soc. Trop. Med. Hyg. 2021, 115, 236–244. [Google Scholar] [CrossRef]
  7. Miguel, D.C.; Brioschi, M.B.; Rosa, L.B.; Minori, K.; Grazzia, N. The impact of COVID-19 on neglected parasitic diseases: What to expect? Trends Parasitol. 2021, 37, 694–697. [Google Scholar] [CrossRef]
  8. Dantas, N.M.; Andrade, L.A.; da Paz, W.S.; Borges, W.N.; Barbosa, V.G.B.; da Hora, D.P.G.; da Silva, C.E.; Carmo, R.F.D.; de Souza, C.D.F.; dos Santos, A.D.; et al. Impact of the COVID-19 pandemic on the actions of the Schistosomiasis Control Program in an endemic area in Northeastern Brazil. Acta Trop. 2023, 240, 106859. [Google Scholar] [CrossRef] [PubMed]
  9. WHO. Schistosomiasis and soil-transmitted helminthiases: Progress report, 2020. Wkly. Epidemiol. Rec. 2021, 96, 585–595. [Google Scholar]
  10. Weerakoon, K.G.A.D.; Gobert, G.N.; Cai, P.; McManus, D.P. Advances in the Diagnosis of Human Schistosomiasis. Clin. Microbiol. Rev. 2015, 28, 939–967. [Google Scholar] [CrossRef] [Green Version]
  11. Hams, E.; Aviello, G.; Fallon, P.G. The Schistosoma Granuloma: Friend or Foe? Front. Immunol. 2013, 4, 89. [Google Scholar] [CrossRef] [Green Version]
  12. Hesse, M.; Piccirillo, C.A.; Belkaid, Y.; Prufer, J.; Mentink-Kane, M.; Leusink, M.; Cheever, A.W.; Shevach, E.M.; Wynn, T.A. The Pathogenesis of Schistosomiasis Is Controlled by Cooperating IL-10-Producing Innate Effector and Regulatory T Cells. J. Immunol. 2004, 172, 3157–3166. [Google Scholar] [CrossRef] [Green Version]
  13. Colley, D.G.; Bustinduy, A.L.; Secor, W.E.; King, C.H. Human schistosomiasis. Lancet 2014, 383, 2253–2264. [Google Scholar] [CrossRef] [PubMed]
  14. Gryseels, B. Schistosomiasis. Infect. Dis. Clin. N. Am. 2012, 26, 383–397. [Google Scholar] [CrossRef]
  15. Scorza, B.M.; Carvalho, E.M.; Wilson, M.E. Cutaneous Manifestations of Human and Murine Leishmaniasis. Int. J. Mol. Sci. 2017, 18, 1296. [Google Scholar] [CrossRef] [Green Version]
  16. Torres-Guerrero, E.; Quintanilla-Cedillo, M.R.; Ruiz-Esmenjaud, J.; Arenas, R. Leishmaniasis: A review. F1000Research 2017, 6, 750. [Google Scholar] [CrossRef]
  17. Marzochi, M.C.D.A.; Marzochi, K.B.F. Tegumentary and visceral leishmaniases in Brazil: Emerging anthropozoonosis and possibilities for their control. Cad. Saude Publica 1994, 10, S359–S375. [Google Scholar] [CrossRef] [Green Version]
  18. Barral, A.; Badaro, R.; Carvalho, E.M.; Barral-Netto, M.; De Jesus, A.R.; Johnson, W.D.; Momen, H.; Almeida, R.; McMahon-Pratt, D.; Pedral-Sampaio, D.; et al. Leishmaniasis in Bahia, Brazil: Evidence that Leishmania amazonensis Produces a Wide Spectrum of Clinical Disease. Am. J. Trop. Med. Hyg. 1991, 44, 536–546. [Google Scholar] [CrossRef] [PubMed]
  19. Follador, I.; Araujo, C.; Bacellar, O.; Araujo, C.B.; Carvalho, L.P.; Almeida, R.P.; Carvalho, E.M. Epidemiologic and Immunologic Findings for the Subclinical Form of Leishmania braziliensis Infection. Clin. Infect. Dis. 2002, 34, e54–e58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Salam, N.; Al-Shaqha, W.M.; Azzi, A. Leishmaniasis in the Middle East: Incidence and Epidemiology. PLoS Negl. Trop. Dis. 2014, 8, e3208. [Google Scholar] [CrossRef] [Green Version]
  21. de Almeida, L.V.; Reis-Cunha, J.L.; Coqueiro-Dos-Santos, A.; Rodrigues-Luís, G.F.; Baptista, R.D.P.; Silva, S.D.O.; de Melo, M.N.; Bartholomeu, D.C. Comparative genomics of Leishmania isolates from Brazil confirms the presence of Leishmania major in the Americas. Int. J. Parasitol. 2021, 51, 1047–1057. [Google Scholar] [CrossRef]
  22. Ruiz-Postigo, J.A.; Jain, S.; Mikhailov, A.; Maia-Elkhoury, A.N.; Valadas, S.; Warusavithana, S.; Osman, M. Global leishmaniasis surveillance: 2019–2020, a baseline for the 2030 roadmap. Wkly. Epidemiol. Rec. 2021, 96, 401–419. [Google Scholar]
  23. Ruiz-Postigo, J.A.; Jain, S.; Madjou, S.; Maia-Elkhoury, A.N.; Valadas, S.; Warusavithana, S.; Osman, M.; Yajima, A.; Lin, Z.; Beshah, A.; et al. Global leishmaniasis surveillance: 2021, assessing the impact of the COVID-19 pandemic. Wkly. Epidemiol. Rec 2022, 97, 575–590. [Google Scholar]
  24. Alvar, J.; Vélez, I.D.; Bern, C.; Herrero, M.; Desjeux, P.; Cano, J.; Jannin, J.; den Boer, M. Who Leishmaniasis Control the WHO Leishmaniasis Control Team Leishmaniasis Worldwide and Global Estimates of Its Incidence. PLoS ONE 2012, 7, e35671. [Google Scholar] [CrossRef] [PubMed]
  25. Scott, P.; Novais, P.S.F.O. Cutaneous leishmaniasis: Immune responses in protection and pathogenesis. Nat. Rev. Immunol. 2016, 16, 581–592. [Google Scholar] [CrossRef]
  26. Stanley, A.C.; Engwerda, C.R. Balancing immunity and pathology in visceral leishmaniasis. Immunol. Cell Biol. 2006, 85, 138–147. [Google Scholar] [CrossRef] [PubMed]
  27. de Araújo, F.F.; Costa-Silva, M.F.; Pereira, A.A.S.; Rêgo, F.D.; Pereira, V.H.S.; de Souza, J.P.; Fernandes, L.O.B.; Martins-Filho, O.A.; Gontijo, C.M.F.; Peruhype-Magalhães, V.; et al. Chemokines in Leishmaniasis: Map of cell movements highlights the landscape of infection and pathogenesis. Cytokine 2021, 147, 155339. [Google Scholar] [CrossRef]
  28. Cox, F.E.G. Concomitant infections, parasites and immune responses. Parasitology 2001, 122, S23–S38. [Google Scholar] [CrossRef] [Green Version]
  29. Petney, T.N.; Andrews, R.H. Multiparasite communities in animals and humans: Frequency, structure and pathogenic significance. Int. J. Parasitol. 1998, 28, 377–393. [Google Scholar] [CrossRef]
  30. Mideo, N. Parasite adaptations to within-host competition. Trends Parasitol. 2009, 25, 261–268. [Google Scholar] [CrossRef] [PubMed]
  31. O’neal, S.E.; Guimarães, L.H.; Machado, P.R.; Alcântara, L.; Morgan, D.J.; Passos, S.; Glesby, M.J.; Carvalho, E.M. Influence of Helminth Infections on the Clinical Course of and Immune Response to Leishmania braziliensis Cutaneous Leishmaniasis. J. Infect. Dis. 2007, 195, 142–148. [Google Scholar] [CrossRef] [Green Version]
  32. Miranda, G.S.; Resende, S.D.; Cardoso, D.T.; Camelo, G.M.A.; Silva, J.K.A.O.; de Castro, V.N.; Geiger, S.M.; Carneiro, M.; Negrão-Corrêa, D. Previous History of American Tegumentary Leishmaniasis Alters Susceptibility and Immune Response Against Schistosoma mansoni Infection in Humans. Front. Immunol. 2021, 12, 630934. [Google Scholar] [CrossRef]
  33. Gregorio, A.W.; Vasconcellos, M.R.; Enokihara, M.M.; Guerra, J.M.; Nonogaki, S.; Tomimori, J. Cutaneous schistosomiasis and leishmaniasis coinfection: A case report. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1781–1783. [Google Scholar] [CrossRef] [PubMed]
  34. Azeredo-Coutinho, R.B.G.; Pimentel, M.I.; Zanini, G.M.; Madeira, M.F.; Cataldo, J.I.; Schubach, A.O.; Quintella, L.P.; de Mello, C.X.; Mendonça, S.C. Intestinal helminth coinfection is associated with mucosal lesions and poor response to therapy in American tegumentary leishmaniasis. Acta Trop. 2016, 154, 42–49. [Google Scholar] [CrossRef] [PubMed]
  35. Paran, Y. Massive Splenomegaly: A Case Report of Visceral Leishmania and Schistosomiasis Co-infection and a Review of Infectious Causes of Massive Splenomegaly. Clin. Case Rep. Rev. 2018, 4, 1–3. [Google Scholar] [CrossRef] [Green Version]
  36. Resende, S.D.; Magalhães, F.C.; Rodrigues-Oliveira, J.L.; Castro, V.N.; Souza, C.S.A.; Oliveira, E.J.; Carneiro, M.; Geiger, S.M.; Negrão-Corrêa, D.A. Modulation of Allergic Reactivity in Humans Is Dependent on Schistosoma mansoni Parasite Burden, Low Levels of IL-33 or TNF-α and High Levels of IL-10 in Serum. Front. Immunol. 2019, 9, 3158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Sokhna, C.; Le Hesran, J.-Y.; Mbaye, P.A.; Akiana, J.; Camara, P.; Diop, M.; Ly, A.; Druilhe, P. Increase of malaria attacks among children presenting concomitant infection by Schistosoma mansoni in Senegal. Malar. J. 2004, 3, 43. [Google Scholar] [CrossRef] [Green Version]
  38. Capron, M.; Béghin, L.; Leclercq, C.; Labreuche, J.; Dendooven, A.; Standaert, A.; Delbeke, M.; Porcherie, A.; Nachury, M.; Boruchowicz, A.; et al. Safety of P28GST, a Protein Derived from a Schistosome Helminth Parasite, in Patients with Crohn’s Disease: A Pilot Study (ACROHNEM). J. Clin. Med. 2019, 9, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Newlove, T.; Machado, P.R.; Alcântara, L.; Guimarães, L.H.; Carvalho, E.M.; Morgan, D.J.; Glesby, M.J. Antihelminthic Therapy and Antimony in Cutaneous Leishmaniasis: A Randomized, Double-Blind, Placebo-Controlled Trial in Patients Co-Infected with Helminths and Leishmania braziliensis. Am. J. Trop. Med. Hyg. 2011, 84, 551–555. [Google Scholar] [CrossRef] [Green Version]
  40. El Ridi, R.; Tallima, H.; Mahana, N.; Dalton, J.P. Innate immunogenicity and in vitro protective potential of Schistosoma mansoni lung schistosomula excretory–secretory candidate vaccine antigens. Microbes Infect. 2010, 12, 700–709. [Google Scholar] [CrossRef]
  41. Reynolds, S.R.; Harn, D.A. Comparison of irradiated-cercaria schistosome vaccine models that use 15- and 50-kilorad doses: The 15-kilorad dose gives greater protection, smaller liver sizes, and higher gamma interferon levels after challenge. Infect. Immun. 1992, 60, 90–94. [Google Scholar] [CrossRef] [Green Version]
  42. Hervé, M.; Angeli, V.; Pinzar, E.; Wintjens, R.; Faveeuw, C.; Narumiya, S.; Capron, A.; Urade, Y.; Capron, M.; Riveau, G.; et al. Pivotal roles of the parasite PGD2 synthase and of the host D prostanoid receptor 1 in schistosome immune evasion. Eur. J. Immunol. 2003, 33, 2764–2772. [Google Scholar] [CrossRef]
  43. Hams, E.; Bermingham, R.; Wurlod, F.A.; Hogan, A.E.; O’Shea, D.; Preston, R.J.; McKenzie, A.N.J.; Fallon, P.G.; Rodewald, H.-R. The helminth T2 RNase ω1 promotes metabolic homeostasis in an IL-33- and group 2 innate lymphoid cell-dependent mechanism. FASEB J. 2016, 30, 824–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Everts, B.; Perona-Wright, G.; Smits, H.H.; Hokke, C.H.; van der Ham, A.J.; Fitzsimmons, C.M.; Doenhoff, M.J.; Van Der Bosch, J.; Mohrs, K.; Haas, H.; et al. Omega-1, a glycoprotein secreted by Schistosoma mansoni eggs, drives Th2 responses. J. Exp. Med. 2009, 206, 1673–1680. [Google Scholar] [CrossRef]
  45. Ittiprasert, W.; Mann, V.H.; Karinshak, S.E.; Coghlan, A.; Rinaldi, G.; Sankaranarayanan, G.; Chaidee, A.; Tanno, T.; Kumkhaek, C.; Prangtaworn, P.; et al. Programmed genome editing of the omega-1 ribonuclease of the blood fluke, Schistosoma mansoni. eLife 2019, 8, 1–27. [Google Scholar] [CrossRef]
  46. Okano, M.; Satoskar, A.R.; Nishizaki, K.; Harn, D.A. Lacto-N-fucopentaose III Found on Schitosoma mansoni Egg Antigens Functions as Adjuvant for Proteins by Inducing Th2-Type Response. J. Immunol. 2001, 167, 442–450. [Google Scholar] [CrossRef] [Green Version]
  47. Okano, M.; Satoskar, A.R.; Nishizaki, K.; Abe, M.; Harn, D.A.A. Induction of Th2 responses and IgE is largely due to carbohydrates functioning as adjuvants on Schistosoma mansoni egg antigens. J. Immunol. 1999, 163, 6712–6717. [Google Scholar] [CrossRef] [PubMed]
  48. Turner, J.D.; Jenkins, G.R.; Hogg, K.G.; Aynsley, S.A.; Paveley, R.A.; Cook, P.C.; Coles, M.C.; Mountford, A.P. CD4+CD25+ Regulatory Cells Contribute to the Regulation of Colonic Th2 Granulomatous Pathology Caused by Schistosome Infection. PLoS Negl. Trop. Dis. 2011, 5, e1269. [Google Scholar] [CrossRef]
  49. Haeberlein, S.; Obieglo, K.; Ozir-Fazalalikhan, A.; Chayé, M.A.M.; Veninga, H.; Van Der Vlugt, L.E.P.M.; Voskamp, A.; Boon, L.; Haan, J.M.M.D.; Westerhof, L.B.; et al. Schistosome egg antigens, including the glycoprotein IPSE/alpha-1, trigger the development of regulatory B cells. PLoS Pathog. 2017, 13, e1006539. [Google Scholar] [CrossRef] [Green Version]
  50. Layland, L.E.; Rad, R.; Wagner, H.; da Costa, C.U.P. Immunopathology in schistosomiasis is controlled by antigen-specific regulatory T cells primed in the presence of TLR2. Eur. J. Immunol. 2007, 37, 2174–2184. [Google Scholar] [CrossRef]
  51. Taylor, J.J.; Mohrs, M.; Pearce, E.J. Regulatory T Cell Responses Develop in Parallel to Th Responses and Control the Magnitude and Phenotype of the Th Effector Populatio. J. Immunol. 2006, 176, 5839–5847. [Google Scholar] [CrossRef]
  52. Bacellar, O.; Lessa, H.; Schriefer, A.; Machado, P.; de Jesus, A.R.; Dutra, W.O.; Gollob, K.J.; Carvalho, E.M. Up-Regulation of Th1-Type Responses in Mucosal Leishmaniasis Patients. Infect. Immun. 2002, 70, 6734–6740. [Google Scholar] [CrossRef] [Green Version]
  53. Faria, D.R.; Gollob, K.J.; Barbosa, J.; Schriefer, A.; Machado, P.R.L.; Lessa, H.; Carvalho, L.P.; Romano-Silva, M.A.; de Jesus, A.R.; Carvalho, E.M.; et al. Decreased In Situ Expression of Interleukin-10 Receptor Is Correlated with the Exacerbated Inflammatory and Cytotoxic Responses Observed in Mucosal Leishmaniasis. Infect. Immun. 2005, 73, 7853–7859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Gaze, S.T.; Dutra, W.O.; Lessa, M.; Lessa, H.; Guimaraes, L.H.; de Jesus, A.R.; Carvalho, L.P.; Machado, P.; Carvalho, E.M.; Gollob, K.J. Mucosal Leishmaniasis Patients Display an Activated Inflammatory T-cell Phenotype Associated with a Nonbalanced Monocyte Population. Scand. J. Immunol. 2005, 63, 70–78. [Google Scholar] [CrossRef] [PubMed]
  55. Cañeda-Guzmán, I.C.; Salaiza-Suazo, N.; Fernández-Figueroa, E.A.; Carrada-Figueroa, G.; Aguirre-García, M.; Becker, I. NK Cell Activity Differs between Patients with Localized and Diffuse Cutaneous Leishmaniasis Infected with Leishmania mexicana: A Comparative Study of TLRs and Cytokines. PLoS ONE 2014, 9, e112410. [Google Scholar] [CrossRef] [PubMed]
  56. Diaz, N.L.; Arvelaez, F.A.; Zerpa, O.; Tapia, F.J. Inducible nitric oxide synthase and cytokine pattern in lesions of patients with American cutaneous leishmaniasis. Clin. Exp. Dermatol. 2006, 31, 114–117. [Google Scholar] [CrossRef] [PubMed]
  57. Diaz, N.L.; Zerpa, O.; Tapia, F.J. Chemokines and chemokine receptors expression in the lesions of patients with American cutaneous leishmaniasis. Mem. Inst. Oswaldo Cruz. 2013, 108, 446–452. [Google Scholar] [CrossRef] [PubMed]
  58. Campanelli, A.P.; Brodskyn, C.I.; Boaventura, V.; Silva, C.; Roselino, A.M.; Costa, J.; Saldanha, A.C.; de Freitas, L.A.R.; de Oliveira, C.I.; Barral-Netto, M.; et al. Chemokines and chemokine receptors coordinate the inflammatory immune response in human cutaneous leishmaniasis. Hum. Immunol. 2010, 71, 1220–1227. [Google Scholar] [CrossRef]
  59. Bafica, A.M.B.; Cardoso, L.S.; Oliveira, S.C.; Loukas, A.; Varela, G.T.; Oliveira, R.R.; Bacellar, O.; Carvalho, E.M.; Araújo, M.I. Schistosoma mansoni antigens alter the cytokine response in vitro during cutaneous leishmaniasis. Mem. Inst. Oswaldo Cruz. 2011, 106, 856–863. [Google Scholar] [CrossRef]
  60. Bafica, A.M.B.; Cardoso, L.S.; Oliveira, S.C.; Loukas, A.; Góes, A.; Oliveira, R.R.; Carvalho, E.M.; Araujo, M.I. Changes in T-Cell and Monocyte Phenotypes In Vitro by Schistosoma mansoni Antigens in Cutaneous Leishmaniasis Patients. J. Parasitol. Res. 2012, 2012, 520308. [Google Scholar] [CrossRef] [Green Version]
  61. Lopes, D.M.; Fernandes, J.S.; Cardoso, T.M.d.S.; Bafica, A.M.B.; Oliveira, S.C.; Carvalho, E.M.; Araujo, M.I.; Cardoso, L.S. Dendritic Cell Profile Induced by Schistosoma mansoni Antigen in Cutaneous Leishmaniasis Patients. BioMed Res. Int. 2014, 2014, 743069. [Google Scholar] [CrossRef]
  62. Lopes, D.M.; de Almeida, T.V.V.S.; Souza, R.D.P.D.; Ribeiro, L.E.V.; Page, B.; Fernandes, J.D.S.; Carvalho, E.M.; Cardoso, L.S. Susceptibility of dendritic cells from individuals with schistosomiasis to infection by Leishmania braziliensis. Mol. Immunol. 2018, 93, 173–183. [Google Scholar] [CrossRef]
  63. Lopes, D.M.; Oliveira, S.C.; Page, B.; Carvalho, L.P.; Carvalho, E.M.; Cardoso, L.S. Schistosoma mansoni rSm29 Antigen Induces a Regulatory Phenotype on Dendritic Cells and Lymphocytes From Patients with Cutaneous Leishmaniasis. Front. Immunol. 2019, 9, 1–13. [Google Scholar] [CrossRef] [PubMed]
  64. Cota, G.F.; Gomes, L.I.; Pinto, B.F.; Santos-Oliveira, J.R.; Da-Cruz, A.M.; Pedrosa, M.S.; Tafuri, W.L.; Rabello, A. Dyarrheal Syndrome in a Patient Co-Infected with Leishmania infantum and Schistosoma mansoni. Case Rep. Med. 2012, 2012, 240512. [Google Scholar] [CrossRef]
  65. Catorze, G.; Alberto, J.; Afonso, A.; Vieira, R.; Cortes, S.; Campino, L. Leishmania infantum/HIV co-infection: Cutaneous lesions following treatment of visceral leishmaniasis. Ann. Dermatol. Venereol. 2006, 133, 39–42. [Google Scholar] [CrossRef] [PubMed]
  66. Ritmeijer, K.; Veeken, H.; Melaku, Y.; Leal, G.; Amsalu, R.; Seaman, J. Davidson Ethiopian visceral leishmaniasis: Generic and proprietary sodium stibogluconate are equivalent; HIV co-infected patients have a poor outcome. Trans. R. Soc. Trop. Med. Hyg. 2001, 95, 668–672. [Google Scholar] [CrossRef]
  67. James, S.L.; Cook, K.W.; Lazdins, J.K. Activation of human monocyte-derived macrophages to kill schistosomula of Schistosoma mansoni in vitro. J. Immunol. 1990, 145, 2686–2690. [Google Scholar] [CrossRef] [PubMed]
  68. McSorley, H.J.; Maizels, R.M. Helminth Infections and Host Immune Regulation. Clin. Microbiol. Rev. 2012, 25, 585–608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Steel, C.; McCarthy, J.S.; Ottesen, E.; Guinea, A. Long-term effect of prenatal exposure to maternal microfilaraemia on immune responsiveness to filarial parasite antigens. Lancet 1994, 343, 890–893. [Google Scholar] [CrossRef]
  70. LaCorcia, M.; Da Costa, C.U.P. Maternal Schistosomiasis: Immunomodulatory Effects with Lasting Impact on Allergy and Vaccine Responses. Front. Immunol. 2018, 9, 2960. [Google Scholar] [CrossRef]
  71. Straubinger, K.; Paul, S.; da Costa, O.P.; Ritter, M.; Buch, T.; Busch, D.H.; Layland, L.E.; da Costa, C.U.P. Maternal immune response to helminth infection during pregnancy determines offspring susceptibility to allergic airway inflammation. J. Allergy Clin. Immunol. 2014, 134, 1271–1279.e10. [Google Scholar] [CrossRef]
  72. Cortes-Selva, D.; Gibbs, L.; Maschek, J.A.; Nascimento, M.; Van Ry, T.; Cox, J.E.; Amiel, E.; Fairfax, K.C. Metabolic reprogramming of the myeloid lineage by Schistosoma mansoni infection persists independently of antigen exposure. PLoS Pathog. 2021, 17, e1009198. [Google Scholar] [CrossRef]
  73. Rheinberg, C.E.; Moné, H.; Caffrey, C.R.; Imbert-Establet, D.; Jourdane, J.; Ruppel, A. Schistosoma haematobium, S. intercalatum, S. japonicum, S. mansoni, and S. rodhaini in mice: Relationship between patterns of lung migration by schistosomula and perfusion recovery of adult worms. Parasitol. Res. 1998, 84, 338–342. [Google Scholar] [CrossRef]
  74. Loker, E.S. A comparative study of the life-histories of mammalian schistosomes. Parasitology 1983, 87, 343–369. [Google Scholar] [CrossRef]
  75. Coelho, P.M.Z.; Mayrink, W.; Dias, M.; Pereira, L.H. Susceptibility to Leishmania mexicana of mice infected with Schistosoma mansoni. Trans. R. Soc. Trop. Med. Hyg. 1980, 74, 141. [Google Scholar] [CrossRef]
  76. La Flamme, A.C.; Scott, P.; Pearce, E.J. Schistosomiasis delays lesion resolution during Leishmania major infection by impairing parasite killing by macrophages. Parasite Immunol. 2002, 24, 339–345. [Google Scholar] [CrossRef]
  77. Yole, D.; Shamala, K.; Kithome, K.; Gicheru, M. Studies on the interaction of Schistosoma mansoni and Leishmania major in experimentally infected Balb/c mice. Afr. J. Health Sci. 2007, 14, 80–85. [Google Scholar] [CrossRef] [Green Version]
  78. Yoshida, A.; Maruyama, H.; Yabu, Y.; Amano, T.; Kobayakawa, T.; Ohta, N. Immune responses against protozoal and nematodal infection in mice with underlying Schistosoma mansoni infection. Parasitol. Int. 1999, 48, 73–79. [Google Scholar] [CrossRef]
  79. Khayeka–Wandabwa, C.; Kutima, H.L.; Nyambati, V.C.S.; Ingonga, J.; Oyoo–Okoth, E.; Karani, L.W.; Jumba, B.; Githuku, K.S.; O Anjili, C. Combination therapy using Pentostam and Praziquantel improves lesion healing and parasite resolution in BALB/c mice co-infected with Leishmania major and Schistosoma mansoni. Parasites Vectors 2013, 6, 244. [Google Scholar] [CrossRef] [Green Version]
  80. Guimarães, L.H.; Machado, P.R.L.; Lago, E.L.; Morgan, D.J.; Schriefer, A.; Bacellar, O.; Carvalho, E. Atypical manifestations of tegumentary leishmaniasis in a transmission area of Leishmania braziliensis in the state of Bahia, Brazil. Trans. R. Soc. Trop. Med. Hyg. 2009, 103, 712–715. [Google Scholar] [CrossRef] [Green Version]
  81. Schubach, A.; Marzochi, M.C.; Cuzzi-Maya, T.; Oliveira, A.V.; Araujo, M.L.; Pacheco, R.S.; Momen, H.; Conceicao-Silva, F.; Coutinho, S.G. Cutaneous scars in American tegumentary leishmaniasis patients: A site of Leishmania (Viannia) braziliensis persistence and viability eleven years after antimonial therapy and clinical cure. Am. J. Trop. Med. Hyg. 1998, 58, 824–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Hassan, M.F.; Zhang, Y.; Engwerda, C.R.; Kaye, P.M.; Sharp, H.; Bickle, Q.D. The Schistosoma mansoni Hepatic Egg Granuloma Provides a Favorable Microenvironment for Sustained Growth of Leishmania donovani. Am. J. Pathol. 2006, 169, 943–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Bertho, A.L.; Santiago, M.A.; Coutinho, S.G. An Experimental Model of the Production of Metastases in Murine Cutaneous Leishmaniasis. J. Parasitol. 1994, 80, 93. [Google Scholar] [CrossRef]
  84. Gomes-Silva, A.; Valverde, J.G.; Ribeiro-Romão, R.P.; Plácido-Pereira, R.M.; Da-Cruz, A.M. Golden hamster (Mesocricetus auratus) as an experimental model for Leishmania (Viannia) braziliensis infection. Parasitology 2013, 140, 771–779. [Google Scholar] [CrossRef]
  85. Dea-Ayuela, M.A.; Rama-Íñiguez, S.; Alunda, J.M.; Bolás-Fernandez, F. Setting New Immunobiological Parameters in the Hamster Model of Visceral Leishmaniasis for In Vivo Testing of Antileishmanial Compounds. Veter.-Res. Commun. 2007, 31, 703–717. [Google Scholar] [CrossRef] [PubMed]
  86. Goto, H.; Lindoso, J.A.L. Immunity and immunosuppression in experimental visceral leishmaniasis. Braz. J. Med. Biol. Res. 2004, 37, 615–623. [Google Scholar] [CrossRef] [Green Version]
  87. Pirmez, C.; Marzochi, M.C.A.; Coutinho, S.G. Experimental canine mucocutaneous leishmaniasis (Leishmania braziliensis braziliensis). Mem. Inst. Oswaldo Cruz. 1988, 83, 145–151. [Google Scholar] [CrossRef] [PubMed]
  88. Karoum, K.O.; Amin, M.A. Domestic and wild animals naturally infected with Schistosoma mansoni in the Gezira Irrigated Scheme, Sudan. J. Trop. Med. Hyg. 1985, 88, 83–89. [Google Scholar] [PubMed]
  89. Moore, D.V.; Sandground, J.H. The Relative Egg Producing Capacity of Schistosoma mansoni and Schistosoma japonicum. Am. J. Trop. Med. Hyg. 1956, 5, 831–840. [Google Scholar] [CrossRef] [PubMed]
  90. Dolabella, S.S.; Coelho, P.M.Z.; Borçari, I.T.; Mello, N.A.S.T.; Andrade, Z.D.A.; Silva, E.F. Morbidity due to Schistosoma mansoniEntamoeba histolytica coinfection in hamsters (Mesocricetus auratus). Rev. Soc. Bras. Med. Trop. 2007, 40, 170–174. [Google Scholar] [CrossRef] [Green Version]
  91. Moore, D.V.; Meleney, H.E. Comparative Susceptibility of Common Laboratory Animals to Experimental Infection with Schistosoma haematobium. J. Parasitol. 1954, 40, 392. [Google Scholar] [CrossRef]
  92. Gentile, R.; Costa-Neto, S.F.; D’Andrea, P.S. A review on the role of the water-rat Nectomys squamipes on the transmission dynamics of mansonic schistosomiasis: A long term multidisciplinary study in an endemic area. Oecol. Aust. 2010, 14, 711–715. [Google Scholar] [CrossRef]
  93. Miranda, G.S.; Rodrigues, J.G.M.; Lira, M.G.S.; Nogueira, R.A.; Gomes, G.C.C.; Silva-Souza, N. Monitoring positivity for Schistosoma mansoni in rodents Holochilus sp. naturally infected. Cien. Anim. Bras. 2015, 16, 456–463. [Google Scholar] [CrossRef]
  94. Bastos, O.d.C.; Sadigursky, M.; Nascimento, M.D.D.S.B.D.; Brazil, R.P.; de Holanda, J.C. Holochilus brasiliensis nanus Thomas, 1897: As an experimental model for filariasis, leishmaniasis and schistosomiasis. Rev. Inst. Med. Trop. Sao Paulo 1984, 26, 307–315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Andrade, M.S.; Courtenay, O.; Brito, M.E.F.; Carvalho, F.G.; Carvalho, A.W.S.; Soares, F.; Carvalho, S.M.; Costa, P.L.; Zampieri, R.; Floeter-Winter, L.M.; et al. Infectiousness of Sylvatic and Synanthropic Small Rodents Implicates a Multi-host Reservoir of Leishmania (Viannia) braziliensis. PLoS Negl. Trop. Dis. 2015, 9, e0004137. [Google Scholar] [CrossRef]
  96. Lima, B.S.; Dantas-Torres, F.; de Carvalho, M.R.; Marinho-Junior, J.F.; de Almeida, E.L.; Brito, M.E.F.; Gomes, F.; Brandao-Filho, S.P. Small mammals as hosts of Leishmania spp. in a highly endemic area for zoonotic leishmaniasis in north-eastern Brazil. Trans. R. Soc. Trop. Med. Hyg. 2013, 107, 592–597. [Google Scholar] [CrossRef]
  97. Teva, A.; Porrozzi, R.; Cupolillo, E.; Pirmez, C.; Oliveira-Neto, M.P.; Grimaldi, G. Leishmania (Viannia) braziliensis-induced chronic granulomatous cutaneous lesions affecting the nasal mucosa in the rhesus monkey (Macaca mulatta) model. Parasitology 2003, 127, 437–447. [Google Scholar] [CrossRef]
  98. Amaral, V.F.; Ransatto, V.A.; Conceição-Silva, F.; Molinaro, E.; Ferreira, V.; Coutinho, S.G.; McMahon-Pratt, D.; Grimaldi, J.G. Leishmania amazonensis: The Asian Rhesus Macaques (Macaca mulatta) as an Experimental Model for Study of Cutaneous Leishmaniasis. Exp. Parasitol. 1996, 82, 34–44. [Google Scholar] [CrossRef]
  99. Torben, W.; Hailu, A. The development of hepatic granulomas in 20 Krad irradiated Schistosoma mansoni cercaria vaccinated grivet monkeys (Cercopithecus aethiops aethiops). Exp. Parasitol. 2007, 117, 376–381. [Google Scholar] [CrossRef]
  100. Else, J.G.; Satzger, M.; Sturrock, R.F. Natural infections of Schistosoma mansoni and S. haematobiumin Cercopithecus monkeys in Kenya. Ann. Trop. Med. Parasitol. 1982, 76, 111–112. [Google Scholar] [CrossRef]
  101. Fairley, N.H. A comparative study of experimental bilharziasis in monkeys contrasted with the hitherto described lesions in man. J. Pathol. Bacteriol. 1920, 23, 289–314. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The immunopathogenesis of Schistosoma and Leishmania infections. The outcomes of both schistosomiasis and tegumentary leishmaniasis are heavily impacted by the host’s immune response, which is the main cause of pathology in these diseases. In the first weeks post-Schistosoma sp. infection, known as the pre-postural phase, a predominantly type 1 immune profile (A) is elicited in response to worm migration and maturation, participating in the control of schistosomula but also causing symptoms such as fever, fatigue, myalgia, and headache. After oviposition begins, hepatotoxic antigens are secreted by schistosome eggs. In response to these antigens, there is a change to a type 2 profile (B). This leads to circumoval granuloma formation, indispensable to properly containing cytotoxic egg antigens and protecting the adjacent tissues from damage; however, granulomas also cause fibrosis and are the main cause of pathology in postural acute schistosomiasis. Subsequently, a regulatory response (reg) takes over, reducing granuloma size and controlling pathology during the chronic phase of infection. On the other hand, these three profiles are highly associated with the outcome of tegumentary leishmaniasis, too, as type 1 responses (A) lead to amastigote killing but also damage neighboring tissue. An unchecked Th1 response is especially present in mucosal leishmaniasis. Meanwhile, type 2 and regulatory responses (B), important for tissue repair, favor amastigote proliferation, and the lack of an appropriate type 1 response can lead to anergy and diffuse leishmaniasis. Therefore, this fine balance between immune response profiles, parasite control, and tissue homeostasis may be considerably impacted during coinfections, possibly contributing to altered morbidity and response to treatment.
Figure 1. The immunopathogenesis of Schistosoma and Leishmania infections. The outcomes of both schistosomiasis and tegumentary leishmaniasis are heavily impacted by the host’s immune response, which is the main cause of pathology in these diseases. In the first weeks post-Schistosoma sp. infection, known as the pre-postural phase, a predominantly type 1 immune profile (A) is elicited in response to worm migration and maturation, participating in the control of schistosomula but also causing symptoms such as fever, fatigue, myalgia, and headache. After oviposition begins, hepatotoxic antigens are secreted by schistosome eggs. In response to these antigens, there is a change to a type 2 profile (B). This leads to circumoval granuloma formation, indispensable to properly containing cytotoxic egg antigens and protecting the adjacent tissues from damage; however, granulomas also cause fibrosis and are the main cause of pathology in postural acute schistosomiasis. Subsequently, a regulatory response (reg) takes over, reducing granuloma size and controlling pathology during the chronic phase of infection. On the other hand, these three profiles are highly associated with the outcome of tegumentary leishmaniasis, too, as type 1 responses (A) lead to amastigote killing but also damage neighboring tissue. An unchecked Th1 response is especially present in mucosal leishmaniasis. Meanwhile, type 2 and regulatory responses (B), important for tissue repair, favor amastigote proliferation, and the lack of an appropriate type 1 response can lead to anergy and diffuse leishmaniasis. Therefore, this fine balance between immune response profiles, parasite control, and tissue homeostasis may be considerably impacted during coinfections, possibly contributing to altered morbidity and response to treatment.
Tropicalmed 08 00383 g001
Table 1. Experimental design of S. mansoni coinfections with dermotropic Leishmania spp.
Table 1. Experimental design of S. mansoni coinfections with dermotropic Leishmania spp.
ReferenceMouse StrainS. mansoni StrainS. mansoni LoadLeishmania sp. and StrainLeishmania sp. LoadTime of CoinfectionChanges in Skin Lesions
Coelho et al.
1980 [75]
Outbred miceLE strain70 cercariaeL. mexicana (L v 22)1 × 108 promastigotes8 weeks post S. mansoni infectionShorter incubation period
Yoshida et al.
1999 [78]
BALB/c &
C57BL/6
Puerto Rico strain20 cercariaeL. major (5ASKH)4 × 107 promastigotes8 weeks post S. mansoni infectionAbsent
la Flamme et al.
2002 [76]
C57BL/6Puerto Rico strain70 cercariaeL. major (FN)5 × 106 promastigotes2 weeks post S. mansoni infectionLarger lesions with delayed healing
Yole et al.
2007 [77]
C57BL/6Kenyan isolate150 cercariaeL. major (NLB-144)3 × 106 promastigotesSimultaneousLarger lesions
Khayeka-Wandabwa et al.
2013 [79]
BALB/cKEN-lab strain70 cercariaeL. major (NLB-144)1 × 106 promastigotesSimultaneousAbsent
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Camelo, G.M.A.; Silva, J.K.A.d.O.; Geiger, S.M.; Melo, M.N.; Negrão-Corrêa, D.A. Schistosoma and Leishmania: An Untold Story of Coinfection. Trop. Med. Infect. Dis. 2023, 8, 383. https://doi.org/10.3390/tropicalmed8080383

AMA Style

Camelo GMA, Silva JKAdO, Geiger SM, Melo MN, Negrão-Corrêa DA. Schistosoma and Leishmania: An Untold Story of Coinfection. Tropical Medicine and Infectious Disease. 2023; 8(8):383. https://doi.org/10.3390/tropicalmed8080383

Chicago/Turabian Style

Camelo, Genil Mororó Araújo, Jeferson Kelvin Alves de Oliveira Silva, Stefan Michael Geiger, Maria Norma Melo, and Deborah Aparecida Negrão-Corrêa. 2023. "Schistosoma and Leishmania: An Untold Story of Coinfection" Tropical Medicine and Infectious Disease 8, no. 8: 383. https://doi.org/10.3390/tropicalmed8080383

Article Metrics

Back to TopTop