Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2024 Feb 6;37(1):e00098-23. doi: 10.1128/cmr.00098-23

Beyond schistosomiasis: unraveling co-infections and altered immunity

Dilhan J Perera 1,2, Cal Koger-Pease 1,2, Kayla Paulini 3, Mohamed Daoudi 2,3, Momar Ndao 1,2,3,4,
Editor: Louisa A Messenger5
PMCID: PMC10938899  PMID: 38319102

SUMMARY

Schistosomiasis is a neglected tropical disease caused by the helminth Schistosoma spp. and has the second highest global impact of all parasites. Schistosoma are transmitted through contact with contaminated fresh water predominantly in Africa, Asia, the Middle East, and South America. Due to the widespread prevalence of Schistosoma, co-infection with other infectious agents is common but often poorly described. Herein, we review recent literature describing the impact of Schistosoma co-infection between species and Schistosoma co-infection with blood-borne protozoa, soil-transmitted helminths, various intestinal protozoa, Mycobacterium, Salmonella, various urinary tract infection-causing agents, and viral pathogens. In each case, disease severity and, of particular interest, the immune landscape, are altered as a consequence of co-infection. Understanding the impact of schistosomiasis co-infections will be important when considering treatment strategies and vaccine development moving forward.

KEYWORDS: Schistosoma, co-infection, immunology, soil-transmitted helminths, Plasmodium, Leishmania, Toxoplasma, Trypanosoma, intestinal protozoa, Mycobacterium, Salmonella, UTI-causing agents, SARS-CoV-2, HIV, cytomegalovirus, hepatitis, arboviruses

INTRODUCTION

Approximately 10% of the world’s population is at risk of infection with Schistosoma spp. parasites, which cause the debilitating disease called schistosomiasis, also known as bilharzia (1). Schistosomiasis is the second most important parasitic disease behind only malaria in terms of global impact (2). Spread through contact with contaminated fresh water, schistosomiasis leads to chronic morbidities that can last for over 40 years and life-threatening health conditions if left untreated (3). Schistosomiasis remains a major epidemiological concern, affecting wide geographical regions, comprising primarily low- to middle-income countries (1, 2). Due to this parasite’s widespread distribution, endemic regions often overlap with areas of endemicity for other pathogens (e.g., malaria, leishmaniasis, and more). Despite geographical overlaps of infection occurrence and numerous works demonstrating the ways Schistosoma helminths modulate the immune system (46), there remains a gap of knowledge in the effects of this parasite on co-infecting pathogens and how co-exposure may affect prognosis (7).

Schistosomiasis is caused by three main species in humans: Schistosoma mansoni, S. haematobium, and S. japonicum. Schistosomiasis pathology varies based on the infecting species; S. haematobium results in urinary schistosomiasis, while S. mansoni and S. japonicum result in intestinal schistosomiasis (3).

Schistosomiasis is commonly found in tropical and subtropical regions of Africa, Asia, the Middle East, and South America. While schistosomiasis transmission occurs in over 70 countries (8), 90% of cases occur in sub-Saharan Africa (9, 10). Within Asia, the disease is common in China, the Philippines, and Indonesia, while in South America, Brazil and Venezuela have the highest rates of infection (8). Regions where this parasite is spread often lack infrastructure and clean water sources, leaving the population vulnerable to infection through contaminated freshwater sources such as rivers, lakes, and ponds (3, 8). Despite efforts to control the spread of schistosomiasis, it remains a significant public health problem worldwide.

SCHISTOSOMA CONTROL CHALLENGES

Individuals with schistosomiasis can be effectively treated with praziquantel (PZQ) which for years has been used to prevent long-term complications of chronic infection (8). PZQ is a helminth paralytic which disrupts the nervous system of adult worms leading to parasite death (11). Despite its safety and efficacy, PZQ alone has not been sufficient to eliminate schistosomiasis. Furthermore, it has been difficult to treat all individuals affected by schistosomiasis through mass drug administration (MDA). There are more cases of infection than doses of drug available, and many of those affected are unable to access treatment, particularly those living in rural areas (12, 13). In addition to the limitations of PZQ, schistosomiasis has been difficult to eliminate for many other reasons. For example, S. japonicum can infect other mammals, allowing the population of animals perpetuating the life cycle to expand beyond humans alone. Furthermore, diagnosis is imperfect, and cases of low-level infections exist, allowing those without overtly debilitating symptoms to go undiagnosed and untreated (14). The development of vaccines for this parasite has been underway since the 1970s (1519), although none have yet been approved for human use, and more importantly, high efficacy has been elusive (20).

The presence of Schistosoma spp. in a particular area depends on the presence of the specific species’ intermediate snail vector(s) (21). As snail habitats have changed over time due to factors such as modifications of waterways [dams and irrigation schemes (22, 23)] and climate change (24), Schistosoma endemicity has and continues to change. One well documented occurrence of such changing endemicity occurred in the Senegal River Basin in the early 1990s. Prior to the building of two dams in the region, intestinal schistosomiasis was not found in the Senegal River Basin, and urogenital schistosomiasis rates were low (25, 26). Two years after the opening of the first dam, an outbreak of intestinal (S. mansoni) schistosomiasis occurred in Richard-Toll, a town on the Senegal River in the lower Senegal River Basin (27, 28). Within 6 years of the dam opening, both S. mansoni and S. haematobium were widespread throughout the Lac de Guiers region, which is connected to the Senegal River at Richard-Toll by an intake canal (25). Differently, the effects of climate change can be seen clearly in the outbreak (24, 29) and potential continuing endemicity (30, 31) of urogenital schistosomiasis in Corsica, France. The outbreak first occurred in 2013, but cases have been linked to the region as recently as 2019 (30, 31). The parasites responsible for these cases are S. haematobium × S. bovis hybrids genetically closest to the Schistosoma of the Senegal River Basin (32); their establishment in Corsica is thought to be due to warming temperatures, the existence of a suitable snail vector already present in the region, and increasing migration/movement between endemic regions of Senegal/West Africa and France (29, 33). These two examples, of the Senegal River Basin and Corsica, highlight the importance of effective schistosomiasis monitoring. Moreover, when considering the constraints of PZQ and untreated infections, these examples illustrate the challenges of eliminating schistosomiasis and shed light on its widespread global presence.

CO-ENDEMICITY

Due to the prevalence of schistosomiasis (over 250 million infections yearly) and its broad geographical distribution (8), co-infections of Schistosoma spp. and other pathogens are common (7). As this parasite is common in low- to middle-income countries, affected individuals may be unaware of their co-infections and/or have limited access to treatment for them. Some common co-infections which affect Schistosoma spp. positive individuals include soil-transmitted helminths (STHs) (34), malaria (35), human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS) (36), and tuberculosis (TB) (37).

Helminth infections are known to be formidable modulators of the host immune response, so it is understandable why many reviews have sought to determine the effects of these helminths during cases of co-infection (3840). Although classified as a helminth, Schistosoma spp. carry unique traits which separate them from the rest. For instance, schistosomes are non-hermaphroditic trematodes which reside in the vascular system, rather than the gut lumen (3). Therefore, it is worth exploring the effect schistosomes, specifically, have on co-infecting pathogens and vice versa. Here, we explore co-infections with major parasitic, bacterial, and viral pathogens which have been reported to be co-endemic with schistosomiasis. The pathogens discussed have been chosen based on frequency, geographical sizes of co-endemic regions, global impact, and the availability of research. Additionally, we discuss the epidemiology and immune mechanisms of co-infections within animal models and, where available, in human data.

SCHISTOSOMA PATHOGENESIS

The prevalence of schistosomiasis is complicated by its elaborate life cycle involving a snail vector. Eggs released into fresh water hatch miracidia, the first larval stage of Schistosoma parasites. Free-swimming miracidia infect different snail vectors in a species-dependent manner: S. mansoni infects Biomphalaria spp., S. haematobium infects Bulinus spp., and S. japonicum infects Oncomelania spp. snails. Within the snail, miracidia become sporocysts and undergo several rounds of asexual reproduction, giving rise to cercariae. Cercariae, the next larval stage of Schistosoma spp., exit the snail and swim through the water. Upon contact with a mammal, the definitive host, they actively penetrate the skin. Once inside the epidermis, cercariae shed their tails, transforming into schistosomula, and reside within the dermis for around 3 days. Penetrating larvae drive the expression of programmed death ligands, PD-L1 and -L2, as well as IL-10 by human dermal dendritic cells (DCs; demonstrated in ex vivo human skin explants), promoting a regulatory phenotype (41). Cercarial penetration may also lead to the development of cercarial dermatitis or “swimmer’s itch.” As the infection continues, schistosomiasis generally presents with mild symptoms and shifts toward T-helper-1 (TH1) skewing immune responses (4) with increases of pro-inflammatory cytokines IL-1, IL-6, and TNFα in peripheral blood (42). During this time, schistosomula make their way into the circulation and travel to the lungs. Within the lungs, the parasite thins and elongates over the course of approximately 7 days, before following the circulation to the liver where the final larval stage matures into juvenile worms. The lung stage of the parasite tends to result in cold and flu-like symptoms including the allergic response, Katayama fever (43), and eosinophilic infiltrates which create lesions visible by radiography (44). In the hepatic portal vein, male and female worms mature and pair, then travel together to the mesentery of the intestines for most Schistosoma spp., except for S. haematobium, where adult worms take residence in the veins around the bladder. Severe pathology takes place as adult worms begin laying eggs. Depending on the species, Schistosoma female worms can lay upward of 300 eggs/per day, half of which are released with excreta, whereas the other portion is embedded within host tissues. Schistosoma eggs are highly immunogenic, and the secretion of soluble egg antigens (SEA) shifts immune responses toward T-helper-2 (TH2) mediators (45). These SEA cause a strong influx of immune cells, triggering inflammation and granuloma development, which is important for host protection and parasite egg egress (46, 47). Chronic schistosomiasis leads to an increase in regulatory T (TREG) cell responses, cirrhosis, and organ fibrosis and failure, if left untreated. S. haematobium infection can also result in genital schistosomiasis (4850) and cervical neoplasia (51), in addition to being a major causative agent of bladder cancer (52).

Although the pathobiology of Schistosoma spp. is generally well described and is summarized in Fig. 1, how this parasite behaves in cases of co-infection has yet to be thoroughly explored.

Fig 1.

Fig 1

Schistosoma pathobiology. Intestinal schistosomiasis is represented by Schistosoma mansoni (Sm) transmitted by Biomphalaria spp. snails, and urinary schistosomiasis is represented by S. haematobium (Sh) transmitted by Bulinus spp. snails. Upon cercarial entry into the skin, schistosomes induce TREG responses. From circulation to adult worm oviposition, immune responses are predominately TH1. As adult worms pair and lay eggs in the intestinal mesentery (Sm) and bladder venus plexus (Sh), soluble egg antigens drive TH2 immunity. Eggs are excreted in feces (Sm) and urine (Sh) to continue the life cycle. Chronic schistosomiasis upregulates TREG responses to protect the host from severe pathology and promote parasite persistence. Illustrative images are not to scale.

SCHISTOSOMA CO-INFECTION BETWEEN SPECIES

Two of the three most common human-infecting Schistosoma species, S. mansoni and S. haematobium, have significantly overlapping endemic areas including much of sub-Saharan Africa and parts of Egypt and the Middle East (21). Given that most Schistosoma infections occur in co-endemic sub-Saharan Africa (8), interspecies co-infection is a significant risk (53). While research is lacking on the epidemiology and morbidity of co-infections at a large scale (country/global), there is a growing body of work at the local scale (village/region), notably in Senegal (5358), Cameroon (23, 59), Nigeria (60), Ghana (61), Kenya (62), Niger (55), and Mali (55, 63). In many of the areas investigated, more than half of infected individuals were co-infected with multiple Schistosoma species (53, 56, 61). Mixed infections were consistently associated with changes in egg elimination patterns, infection intensity, and morbidity.

One common indicator of co-infection with S. mansoni and S. haematobium is ectopic egg elimination, which refers to S. haematobium eggs eliminated in the stool and/or S. mansoni eggs eliminated in the urine (Fig. 2). This phenomenon can sometimes be attributed to unusual worm localization or “spillover” from typical egg laying sites in mono-infections, particularly in the case of high parasite burdens (23). More commonly, however, ectopic egg elimination is associated with co-infections and occurs due to heterospecific mating between different Schistosoma species (23, 58, 61). When worms of both species are present in an individual, heterospecific mating (when a male of one species couples with a female of another) can occur in the hepatic portal vein (64). The species of the male schistosome will determine where the couple migrates and lays eggs. S. haematobium schistosomes have a higher male to female ratio (23) than S. mansoni schistosomes and an increased ability to “steal” females from already established pairings (65). These differences explain the higher frequency of S. mansoni eggs found in urine compared to S. haematobium eggs found in stool (23, 53, 58, 59, 61). While ectopic egg elimination has been documented as early as the late 19th century (66), it remains poorly examined and as such it is difficult to accurately estimate its prevalence. The effect of ectopic eggs on the host is also not fully understood in the context of co-infection. However, given that eggs drive schistosomiasis pathology, their presence in “non-traditional” locations could have consequences for infection morbidity, as observed in ectopic mono-infections (67).

Fig 2.

Fig 2

Schistosoma mansoni (Sm) and S. haematobium (Sh) co-infection. In co-infections with Sm and Sh, ectopic egg elimination occurs, with both Sm and Sh eggs found in urine and, to a lesser extent, in feces. Male worms determine the residency of paired couples. Sm male and Sh female couplings (M) are more common, these pairs reside in the venous plexus similar to Sh couples; Sm couples reside within the intestinal mesentery. Co-infection reduces the risk of liver pathology but increases parasite burdens, urogenital pathology, and TH2 immune responses. Illustrative images are not to scale.

Multiple studies have found that higher infection intensities occur in co-infection vs mono-infection with Schistosoma species. An association between high infection burdens and mixed infections has been observed in studies in Niger (68); Niger, Mali, and Senegal (55); the Senegal River Basin (53); Ghana (61); and Cameroon (23). Taken together, these studies suggest a notable effect of co-infection on infection intensity, which is an important consideration for surveillance, treatment, and control efforts. Thus far, multiple studies have shown that PZQ treatment efficacy is equally effective in mono-infections and co-infections (68, 69). However, because co-infections were generally higher burden infections, the dose of PZQ may not be as effective for clearance as in lower burden mono-infections (55). Additional research is needed to fully understand the association between co-infection and higher infection burden, and whether this relationship affects PZQ treatment efficacy.

There have been notable differences in morbidity observed in co-infections with S. mansoni and S. haematobium compared to single-species infections. Specifically, hepatic and splenic pathologies appear to decrease in co-infections, while bladder pathologies tend to increase. In Cameroon, it was found that hepatomegaly and splenomegaly (assessed by physical exam) were less common in co-infections than in S. mansoni mono-infections (23). Similarly, in Mali, ultrasonographic examination showed a decreased risk for abnormal liver imaging for co-infected vs S. mansoni mono-infected [standardized for level of infection (63)]. It was also found that the risk of hepatomegaly was higher in individuals mono-infected with either S. mansoni or S. haematobium than in co-infected individuals; in contrast, bladder pathology was increased in co-infections over S. haematobium mono-infections (63). It is hypothesized that this effect can be attributed to S. haematobium males outcompeting S. mansoni males for mates, resulting in a greater number of S. mansoni females directed to the bladder venous plexus instead of the mesentery of the intestines (65). This shift in worm pair location would lead to preferential egg entrapment and pathology in the bladder versus the liver. Interestingly, one study in Kenya found that bladder pathology was decreased in co-infections compared to S. haematobium mono-infections (62), which may be a result of different sampling techniques or point to population differences, both in terms of humans and parasites. As schistosomiasis epidemiology is highly focal, morbidity varies between locations. This is exemplified by a study in the Senegal River Basin, which found a strong geospatial cluster for severe hepatic fibrosis within the co-endemic region (57). Overall, it appears that the hallmark pathology of S. mansoni is decreased in co-infection, while the pathology of S. haematobium is increased. Still, broader investigations are needed to fully understand the effect of co-infection on morbidity throughout large co-endemic regions.

To date, minimal research has been conducted on the immunology of S. mansoni and S. haematobium co-infections. In general, schistosome immunology research is heavily biased toward S. mansoni, in part due to the lack of a rodent model that accurately recapitulates the life cycle of S. haematobium in humans (70). While Schistosoma immune responses share many similarities, there may be underappreciated species-specific effects, including immunopathology and cancers of the bladder and genital regions, and a lower incidence of hypersensitivity reactions (e.g., cercarial dermatitis and Katayama syndrome) observed in S. haematobium infections (70). When whole blood from individuals living in a co-endemic region was cultured with either S. haematobium or S. mansoni antigens, cytokine responses were similar, and there was an inverse correlation between cytokine responses and Schistosoma infection intensities. Strong TH1 responses were observed in uninfected samples, an intermediate profile was found in single infections and light infections, and a TH2 bias (and lower general response) was found in mixed and heavy infections. Unfortunately, due to the confounding effect of infection intensity, it is unclear whether this gradient in cytokine response is due to the co-infection or the higher parasite burdens seen in that population (56).

Further complicating the picture, co-infections with multiple schistosome species can also lead to species hybridization. Schistosoma hybridization has important implications for host range (vertebrate and snail), pathology, and epidemiology (71, 72). During co-infection, when heterospecific pairing occurs, there is a possibility for parthenogenesis (asexual reproduction resulting in eggs containing only the female’s genome), hybridization (full mixture between the parental genomes), and introgression (transfer of genes/chromosomal regions between species through backcrossing) (64, 73). The likelihood of hybridization between two species is related to their phylogenetic distance, with intraclade hybridizations more frequently observed and established in the literature (71). For instance, hybrids identified in West Africa as well as Corsica, France consist of S. haematobium and the bovine species S. bovis (71). It was previously believed that phylogenetically divergent species, such as S. haematobium and S. mansoni, could not hybridize and that eggs from these heterospecific pairings would be purely parthenogenic. However, over the past decade, there has been growing evidence of hybridization between these two species in humans (7476). Ectopic egg elimination is one effect of interspecific pairings, but given the lack of genotyping evidence, it is not known to what extent those eggs are parthenogenic or hybridized. Hybridization can also affect the host range of the parasite, leading to changes in zoonotic transmission dynamics as seen with S. haematobium × S. bovis hybrids (71). It is also important to consider the effects of hybridization on morbidity profiles, PZQ efficacy, and vaccine development. With continuing changes in human migration, climate, and irrigation and development, interactions between different species of schistosomes and their hosts are likely to continue to shift and increase (71, 72). To keep pace with this rapidly changing landscape, it is imperative to implement more effective surveillance efforts for both co-infections and hybridization events and to investigate these phenomena in the laboratory.

Co-infections with multiple schistosome species and infections with hybridized species are currently underrecognized and understudied. On a local level, multiple studies in S. mansoni and S. haematobium co-endemic areas have found that co-infection is the norm (53, 56, 61) leading to trends of increased worm burdens (23, 53, 55, 61, 68) and ectopic egg elimination (23, 53, 58, 59, 61). Morbidity associated with S. haematobium appears to increase in co-infection, while S. mansoni-associated morbidity decreases (23, 63); future work should assess if these co-infections result in increased manifestations of genital schistosomiasis. These observations need to be validated in larger studies in more locations, and a greater understanding of the effects of co-infection and hybrid infection on immunology, morbidity, diagnosis, treatment, and transmission is necessary.

SOIL-TRANSMITTED HELMINTHS

Soil-transmitted helminths are a group of intestinal parasites infecting up to a quarter of the world’s population. STHs cause neglected tropical diseases which primarily affect impoverished communities with limited access to sanitation in tropical and subtropical regions, especially in sub-Saharan Africa, South America, and Asia. There are three main types of STH: Ascaris lumbricoides (roundworm), Trichuris trichiura (whipworm), and hookworms (e.g., Necator americanus and Ancylostoma duodenale). Additionally, Strongyloides stercoralis (threadworm) is an STH but is often excluded from analyses and control efforts due to differences in diagnosis and treatment compared to the other three STHs (77). Light infections with STHs are typically asymptomatic, but heavier infections can cause a variety of symptoms including diarrhea and abdominal pain, anemia, nutrient deficiency, malnutrition, growth impairment, and intestinal blockage (77, 78). Effective deworming treatments exist for STHs, typically albendazole and mebendazole for the three classic STHs, and ivermectin for S. stercoralis (77).

Immune responses to STH infections are broadly similar, characterized primarily by systemic and local type-2 responses (78, 79). In the gut, the “weep and sweep” response predominates, which is defined by increased mucous production and smooth muscle contraction (79). The cytokine response includes early production of IL-25 and IL-33, followed by long-term production of IL-5, IL-13, IL-4, and IL-10 (80). Chronic infection with STHs requires a degree of immune tolerance (characterized primarily by IL-10), allowing the worm to survive but protecting the host from excessive immunopathology. This semi-tolerant state is known as a modified TH2 response (81).

Generally, STH and schistosomiasis endemicity patterns are similar; however, because schistosomes require the presence of freshwater bodies and snail hosts, their epidemiology is more focal than STHs, which can occur anywhere suitably moist soil and fecal contamination intersect (82). STHs and schistosomiasis are both heavily associated with poor sanitation and are targeted by the World Health Organization (WHO) global Water, Sanitation, and Hygiene (WASH) program and MDA efforts (83). While both diseases are caused by macroparasitic worms, differences in their niches within the host result in differences in the host immune response. The characteristic helminth “weep and sweep” response does not apply to blood vessel-dwelling schistosomes; likewise, egg-related granulomatous immunopathology is not typical of STH infections. Nevertheless, both types of worms induce significant TH2 responses, albeit in differing contexts, and modulate the immune response toward a regulatory profile (84).

Research into immune responses in Schistosoma-STH co-infections has seen an uptick in recent years, but large gaps in understanding remain. A 2008 review on the topic found several studies on animal models of co-infection as well as epidemiological patterns, but few papers investigated their immune responses (34).

Co-infections with STHs and schistosomes are well established to have different infection burdens than mono-infections (higher or lower, depending on various factors), as elaborated in reviews (7, 34). Within S. mansoni and Strongyloides venezuelensis (a rodent helminth used to model St. stercoralis) co-infections, St. venezuelensis infection burdens differed, as did immune responses in the lungs and small intestines, based on the timing of one infection relative to the other. When mice were simultaneously co-infected with S. mansoni and St. venezuelensis, the infection burden was equal to that of St. venezuelensis mono-infection. However, when infected with S. mansoni prior to St. venezuelensis, mice had significantly lowered St. venezuelensis burdens (up to 92% lower), regardless of whether the secondary infection was 2, 4, or 14 weeks after the first. This suggests that the life stage of the schistosomes may be independent of conferred Strongyloides immunity. Given that schistosome-induced immune responses in mice follow a TH1-TH2-regulatory trajectory, it is somewhat surprising that both early and chronic schistosome infections would be protective against Strongyloides. In the lungs and small intestine, the characteristic TH2 responses of St. venezuelensis infection was not affected by the background Schistosoma infection; St. venezuelensis infection induced local production of IL-4 and IL-5, eosinophilia, and increased serum IgE. The TH1 and regulatory responses, however, did differ, depending on when the secondary infection occurred. St. venezuelensis co-infection during acute schistosomiasis resulted in lower IFNγ and IL-10 in the lungs and intestines compared to co-infection during chronic schistosomiasis (85).

While Strongyloides burden varied depending on the time of S. mansoni co-infection, St. venezuelensis had no effect on S. mansoni worm burden, regardless of the timing of co-infection. That said, the immune response and pathology in the liver were altered. When compared to Schistosoma-mono-infected mice, those co-infected simultaneously with Strongyloides showed a greater TH1-skewed cytokine profile in the liver, while those infected 2 weeks after Schistosoma had a greater TH2-skewed profile. When Strongyloides was introduced 4 weeks after schistosomiasis, cytokine production increased, but there were no discernible changes in profile. Notably, liver granuloma size at 15 weeks post-S. mansoni infection was significantly increased in mice co-infected with St. stercoralis simultaneously, 2 weeks, or 4 weeks after S. mansoni infection compared to S. mansoni-mono-infected mice (10%–15% increase in inflamed liver tissue per field of view). Markers of liver injury were also significantly increased in mice infected simultaneously and those infected with Strongyloides 2 weeks post S. mansoni. In contrast, mice infected with St. venezuelensis during chronic schistosomiasis (14 weeks post S. mansoni infection) had no significant changes in their cytokine profile or liver pathology compared to those which were S. mansoni mono-infected (86). It is possible that the presence of another pathogen during the inflammatory stages of early schistosomiasis exacerbated pathology, whereas the immunoregulatory stages of chronic schistosomiasis were able to prevent changes to pathology. These results highlight the complexity of the immune response to co-infection and how the immune background of the individual can affect their response to additional infectious insults. While it is important to consider the temporal dynamics of co-infection, in human populations, especially in endemic areas, information regarding timing of infection is exceedingly rare. It is also worth noting that it is typical for individuals to be exposed to STHs earlier in life and schistosomes later, as water contact increases through childhood (34), calling into question the translational relevance of the infection pattern applied here.

In addition to changes in worm burdens, co-infections can result in pathology changes. In mice, co-infection with Heligmosomoides polygyrus (model organism for hookworm) and S. mansoni resulted in lower schistosome-induced hepatic egg pathology (7, 87). In contrast, studies investigating the effect of whipworm infection on schistosomiasis egg pathology have found that this co-infection led to significantly increased pathology (7, 88, 89). One recent study compared schistosomiasis in baboons already chronically infected with T. trichiura to those mono-infected with S. mansoni. The authors found that while worm and egg burdens were similar between the groups, the co-infected group had significantly larger liver granulomas; the average size was nearly double that of the mono-infected group. Additionally, RNA sequencing analysis showed enrichment in pathways predictive of liver damage and hepatic injury in the co-infected group compared to the mono-infected group (89). As in the S. mansoni and St. venezuelensis co-infection experiments, these results suggest that even in cases where parasite burden is not influenced by co-infection, changes in pathology should still be considered and may be highly significant.

Naturally, immunomodulatory pathogens like helminths have been shown to change reactivity to antigen, and the same effect can be observed when they co-infect. This has been demonstrated in samples from individuals co-infected with a hookworm infection and either A. lumbricoides or S. mansoni or both (90). Peripheral blood mononuclear cells (PBMCs) from co-infected individuals showed significantly lower TNFα, IFNγ, and CXCL10 responses upon hookworm antigen stimulation compared to those infected with hookworm alone. In contrast, co-infected individuals had higher hookworm-antigen-specific antibody responses (IgG subclasses and IgE). Additionally, there was a strong negative correlation between fecal hookworm egg counts and IL-10 production in mono-infected individuals which was not present in co-infected individuals. Overall, these data suggest that co-infection dampens hookworm-specific TH1 responses; however, it is impossible to determine whether this effect is due to the addition of A. lumbricoides or S. mansoni as the groups were not separated (90).

Another immune modulation seen in STH and schistosome infection is changes in the level of trefoil factor family proteins (TFF1, 2, and 3), a family of glycoproteins involved with tissue repair at mucosal surfaces (91). In the gut, TFFs are secreted by goblet cells, and TFF2 and TFF3 are most prevalent (91). TFF2 levels were significantly higher in hookworm-mono-infected individuals, especially female participants, compared to endemic and non-endemic controls. This shift was not seen in S. mansoni-mono-infected or in co-infected individuals. Conversely, TFF3 levels were higher in S. mansoni-mono-infected but not hookworm-mono-infected or -co-infected participants. In vitro analysis showed that treatment with recombinant human TFF2, but not TFF3, could suppress an artificially induced high-inflammatory state. Thus, TFF2 may be used in hookworm-induced immunomodulation (91) which may be abrogated during S. mansoni co-infection. This work suggests that TFFs have a role in anti-helminth immunity, but more studies will be needed to fully elucidate that role and how co-infection may change it.

Given the increases of IgE in helminth infections and the links between helminths and allergy (81), another area of investigation is the effect of co-infection on this association. Simultaneous treatment of PZQ and albendazole in a region of Uganda co-endemic for schistosomiasis and hookworm resulted in increased anti-Schistosoma adult worm IgE production which was associated with a reduced susceptibility to S. mansoni reinfection. However, the same immunity was not developed for hookworm (92). In fact, IgE specific for hookworm adult worm antigen and S. mansoni egg antigen remained unchanged or significantly decreased. Yet, despite these decreases, histamine release in response to adult worm antigens of both helminths was found to increase after treatment. Additionally, histamine release in response to environmental allergen house dust mite (HDM) increased after treatment (93). This finding suggests that helminth co-infections may suppress histamine release and, in turn, allergy. This hypothesis was supported when effective treatment of S. mansoni and hookworm led to enhanced levels of HDM Der p1-specific IgE, a risk factor for the development of allergic disorders (94). These studies present preliminary evidence supporting the hygiene hypothesis in hookworm/S. mansoni co-infections; albeit, there are not yet sufficient data to say that co-infection with these two worms is protective against allergy.

STH infections are one of the most common infections worldwide and are prevalent nearly everywhere schistosomiasis occurs. The interactions between these worms are complex and warrant further study. Co-infection with S. mansoni following St. venezuelensis infection resulted in lowered St. venezuelensis burden, but the opposite did not hold true (85, 86). Although the presence of STH infection (St. venezuelensis or hookworm) was not shown to decrease schistosome burden in the studies discussed here, there was a notable decrease in liver pathology observed in co-infections (86, 87). This can be logically attributed to the immunoregulatory effects of STH infection. In addition to infection burden and pathology, co-infection seems to shift cytokine dynamics (90), levels of TFFs (91), and histamine release (92, 93). These observations are preliminary and should be validated in further studies. There is also a significant lack of research into the effects of S. haematobium and S. japonicum co-infection with STHs. It is important to understand the immune responses and pathology changes that occur during these co-infections, given how commonly they co-infect.

PLASMODIUM

Plasmodium spp. are protozoan parasites responsible for malaria. There are five species of Plasmodium which infect humans: notably, Plasmodium falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi (35). The WHO estimated 247 million cases of malaria and over 600,000 deaths in 2021 (95). The most common malaria symptom is fever, which occurs primarily in children under the age of 5. Treatment varies depending on the species; however, artemisinin-based combination therapies are the favored choice with over 98% cure rates (35). One vaccine has recently been approved and recommended for malaria prevention in children, RTS,S/AS01, a recombinant vaccine containing the P. falciparum circumsporozoite protein and a hepatitis B surface protein and providing 30% protection from severe disease, although another, R21/Matrix-M, is showing promising results in clinical trials (96).

Over 40 species of the Anopheles mosquito can transmit malaria (35). During a bloodmeal, mosquitoes inject sporozoites into the host which infect liver cells. The sporozoites form a schizont within the liver cells which burst, allowing merozoites to escape and infect erythrocytes. Individuals living in malaria prevalent regions typically undergo a gradual and partial development of immunity over several years, which provides low levels of protection against the disease. This protection is believed to be due to anti-sporozoite and anti-merozoite IgG antibodies (35). Additionally, IFNγ production increases parasite phagocytosis and, thus, increases parasite clearance (97).

Helminth and Plasmodium co-infection is common due to their geographical overlap. Schistosoma and Plasmodium are co-endemic to sub-Saharan Africa, Asia, and South America (98). Varying rates of co-infection have been reported, as high as 34% in sub-Saharan Africa (99) and more recently 2.8%, 8.36%, and 8.7%, in Ethiopia (100), Ghana (101), and Cameroon (102), respectively. Since most studies looked at the prevalence of co-infection in children, it is important to note that the rate of pregnant women co-infected with S. haematobium and P. falciparum was 15.2% in Cameroon (103).

In Cameroon and Uganda, school age children with Schistosoma and Plasmodium co-infections led to reduced Plasmodium parasite burdens and in Cameroon, a reduced level of Schistosoma infection when compared to children with mono-infections (102, 104). It was hypothesized that this was a consequence of the early TH1 response elicited by schistosomiasis which helped control the Plasmodium infection (104), though it was not determined which infection came first. In most studies, the rate of anemia was higher in co-infected patients compared to Schistosoma or Plasmodium mono-infections (100103, 105, 106) except in Ghana where moderate levels of anemia only occurred in 5% of co-infected patients compared to schistosomiasis (21.7%)- or malaria (17.8%)-mono-infected patients with no statistical difference in hemoglobin levels between groups (107). In Cameroon, the most common symptoms of Schistosoma and Plasmodium co-infection in children were hematuria, microcytosis, and fever (102). High levels of hematuria were also seen in children in Nigeria where 57.1% of children were infected with S. haematobium and P. falciparum. In this population, 63.8% of co-infected children had hematuria compared to 52.2% of the S. haematobium group and 43.7% of the P. falciparum group (108).

When considering the immunopathology of co-infection, Schistosoma and Plasmodium co-infection led to an increase of anti-Plasmodium (5%–15%) and anti-Schistosoma (6%–22%) IgG, an increase in anti-Plasmodium IgG1 (0.43–0.9 pg/mL), IgG2 (0.25–0.87 pg/mL), and IgG3 (0.80–1.53 pg/mL), an increase in IL-4 (40% for co-infection vs 3.3% for mono-infection), and a decrease in γδ T cells (109111). It was hypothesized that the co-infected children have a helminth-induced TH2-skewed immune response which could explain the increase in anti-malaria antibodies, leading to lower P. falciparum parasite densities (110).

In non-human primates, chronic schistosomiasis protected animals from severe P. knowlesi infection, and they had reduced levels of Plasmodium parasite (81.25% vs 25% protection). Interestingly, Schistosoma- and Plasmodium-co-infected animals had lower levels of IFNγ compared to Plasmodium-only infections and compared to PZQ-treated animals. There also appeared to be lower levels of IL-10 and TNFα after P. knowlesi infection in the baboons with schistosomiasis compared to those without (112). In a multi-dose P. knowlesi infection model in baboons with and without prior schistosomiasis infection, anti-P. knowlesi IgG and IgG1 levels and CD4+ memory T cells were found to be lower in the co-infected animals compared to those that only had P. knowlesi infections (113). This contradicts the epidemiological data, showing that humans had higher IgG levels when co-infected with schistosomiasis (109, 110). While it is unclear how malaria protection was mediated in this model, monocyte levels were higher in the schistosomiasis- and P. knowlesi-co-infected baboons compared to the P. knowlesi-mono-infected baboons (113).

In C57BL/6 mice infected with S. japonicum and P. berghei, co-infected mice had lower levels of IFNγ but higher levels of IL-4, IL-5, IL-13, and TGFβ when compared to P. berghei single infections. The co-infected mice also had lower levels of splenic CD4+ and CD8+ T cells but higher levels of TREG cells than the Plasmodium-only infected group (114). S. mansoni and P. yoelii or P. berghei co-infection in BALB/c, C57BL/6 mice, and CBA/J mice had significantly lower levels of Plasmodium in liver cells of co-infected mice compared to mono-infected mice. Similarly, when mosquitos were allowed to feed on co-infected and Plasmodium-only infected mice, there was less Plasmodium in the mosquitos that fed on mice with S. mansoni infection compared to those that fed on mice without (115). In addition, liver burden protection conferred by schistosomiasis was abolished in IFNγ−/− and IL-4−/− mice, suggesting that these cytokines play a protective role in Plasmodium infection despite the reduction of IFNγ in co-infected animals.

Of particular interest is the role co-infection with malaria and schistosomiasis plays in parasite treatment. In Ethiopia, the malaria treatment artemether-lumefantrine, cured all cases of schistosomiasis in malaria-co-infected patients (116). Similarly, in the Democratic Republic of Congo, children co-infected with Plasmodium and S. mansoni were treated with artesunate-amodiaquine; S. mansoni was cured in 74.4% of these children and partially reduced in 22% (117). While it is unclear how these anti-malaria drugs kill schistosomes, both these parasites share certain characteristics such as residing in the blood, hemoglobin digestion (118, 119), and susceptibility to reactive oxygen species (ROS) and calcium signaling disruption (120, 121). These shared features suggest that anti-malaria drugs which act through these mechanisms may also target schistosomes, and therefore, it may be feasible to clear both parasitic infections with a single treatment.

Although there is great variability in the incidence rate of Schistosoma and Plasmodium co-infection, these rates can be as high as 34% (99) and 57.1% (108), making co-infection an important consideration when studying schistosomiasis. Overall, co-infection appears to lead to a decrease in malarial parasite load (102, 104, 115), but an increase in anemia (100103, 105, 106) is depicted with relevant immunological changes in Fig. 3. The immunopathology of co-infection is more complex where TH2 responses seem to be favored over TH1 responses in both humans and mice; however, non-human primate studies show decreases in both TH1 and TH2 responses (112, 113). Still, studies show that malaria treatments have been successful in treating co-infections (116, 117). With the development of newly approved malaria vaccines, it will be of interest to determine their effects on co-infections of malaria and schistosomiasis.

Fig 3.

Fig 3

Proposed immunological effects of schistosomiasis and malaria co-infection. In recent epidemiological studies and in mouse co-infection models, schistosomiasis leads to an increase in TREG cells, an increase in anti-malaria antibodies and in IL-4 (TH2-skewed response) which appear to reduce the levels of Plasmodium and decrease pathology. However, co-infection increases the risk of anemia. More research must be done to clarify the effects of other aspects of the immune response since they remain unclear.

LEISHMANIA

Leishmania are intracellular protozoan parasites, which cause a range of diseases depending on the species (122). Cutaneous leishmaniasis (CL) is the most common and least severe form of the disease, defined by skin lesions. CL can be caused by a plethora of Leishmania species, including Leishmania major, L. tropica, L. aethiopica, L. mexicana, L. amazonensis, L. braziliensis, L. guyanensis, and more recently described hybrid parasites (122, 123). There is an estimated 700,000–1.2 million new cases of CL globally, per year (122). Visceral leishmaniasis (VL) is considered the most severe form of the disease which is characterized by enlargement of the liver and spleen and causes anemia, internal bleeding, and immunosuppression which can be fatal when untreated. VL is caused by L. donovani and L. infantum with approximately 50,000 cases, annually (122). Despite this, no vaccine has been approved for human leishmaniasis (124, 125). Treatment varies greatly depending on the species and region of the infection, but some options include amphotericin, pentavalent antimonials, miltefosine, paromomycin, and thermotherapy (122).

The Leishmania life cycle begins when a host is bitten by a sandfly (the vector) and is injected with a promastigote, a long-flagellated eukaryotic cell. Injected promastigotes are phagocytized by neutrophils and macrophages and then transform into non-flagellated, circular cells, called amastigotes. Amastigotes replicate within the macrophage and go on to infect other macrophages. The life cycle is completed when a sandfly takes a bloodmeal from an infected host, and within the sandfly, amastigotes transform back into promastigotes (126). Depending on the species, Leishmania can have a variety of animal reservoirs, except for L. donovani which only infects humans (122).

Leishmania infection can be successfully cleared by a TH1 immune response where IFNγ and TNFα elicit nitric oxide (NO) and ROS, both of which are released in infected macrophages, killing Leishmania amastigotes (127). This TH1 response can elicit lifelong protection against Leishmania re-infection following treatment or asymptomatic infections (128).

Since Schistosoma and Leishmania are co-endemic to many countries within Africa (e.g., Sudan), South America (e.g., Brazil), the Middle East, and Asia (98, 122), it is important to consider the potential of co-infection. Surprisingly, few cases of co-infection have been described.

In Brazil, the occurrence of S. mansoni and CL co-infection is 16.7% and 43.2% depending on the region, and patients with a history of CL were two times more likely to be infected with S. mansoni when compared with those who did not have a history of CL (129). S. mansoni infection increased CL patient healing time after antimony treatment despite co-infected patients having smaller lesions (280 mm2) than CL-only infected patients (400 mm2). This could be due to a TH2-skewed response shown by elevated levels of IgE (1,578 vs 427 IU/mL) and IL-5 (364 vs 184 pg/mL) in co-infected patients (129). Furthermore, Leishmania appeared to alter immune responses to S. mansoni since co-infected patients had lower levels or occurrence of eosinophilia (34% vs 51%), CCL17 (233.5L vs 341.0 pg/mL), and IL-17 (2% vs 18.5%), factors (129) that are usually high in S. mansoni infection (127, 130, 131). Since both population-based studies were conducted in Brazil, it would be important to conduct research in other geographical locations where these parasites are endemic to see if these phenomena are transferrable.

In VL cases, gastrointestinal pathology is rare but may be possible in VL and S. mansoni co-infection (122). A case report of a patient with visceral leishmaniasis, anti-leishmanial antibodies, and chronic diarrhea described a failure to resolve the diarrheal disease after amphotericin B treatment and resolution of visceral disease. S. mansoni eggs were identified in the patient’s stool, and following PZQ treatment and a second round of amphotericin B, the diarrhea is fully resolved (132). Similarly, another patient presenting with sporadic diarrhea tested positive for S. mansoni and Leishmania. However, instead of visceral disease, this patient presented with cutaneous lesions on the abdomen and elbow. Praziquantel and meglumine antimoniate were able to resolve both infections (133).

Experimental mouse models of Schistosoma and Leishmania examined the immunopathology of co-infection. When mice were infected with S. mansoni followed by L. major, lesions were larger, and CL resolution was delayed compared to L. major infection alone (134). This contrasts with population-based studies in humans which showed smaller lesions in co-infected individuals yet a similar increased healing time (129). This delayed healing may be a consequence of the decrease in IFNγ, NO, and TNFα in the popliteal lymph nodes of co-infected mice vs L. major-only infected mice, suggesting an impaired TH1 response resulting in decreased macrophage killing of amastigotes. In parallel, IL-4 was upregulated in co-infected mice which suggests an upregulated TH2 response (134). Similarly, when mice were infected with L. donovani and S. mansoni, leishmanial parasite burden increased over time (2720 LDU; Leishman-Donovan units) when compared to L. donovani single infection (82 LDU) (135). Just like the L. major co-infections (134), L. donovani-co-infected mice showed elevated IL-4 compared to singularly infected mice. They also showed increased IL-10 and decreased IFNγ, but these levels equated over time. Moreover, the S. mansoni granuloma appeared to be a favorable growth environment for L. donovani in mice, demonstrated by increased levels of amastigotes and decreased expression of NO in co-infected mice compared to mice infected with L. donovani alone (135).

Since disease pathology was altered in Schistosoma and Leishmania co-infection, it is important to consider if response to treatment is also altered. In BALB/c mice co-infected with S. mansoni and L. major, both lesion size and parasite burden had a greater reduction in mice that were treated with antimonials and PZQ compared to treatment with only one of the two drugs (136). Although there have been no signs of drug interaction when using combined chemotherapy in mice, some liver toxicity was noted at the beginning of treatment (136). Toxicity of combined chemotherapies should be further examined in humans.

Unfortunately, little is known about the rate of Schistosoma and Leishmania co-infection except in Brazil, where rates are estimated to be as high as 43.2% depending on the region (129). It is not known if these rates are representative of other countries where co-infection occurs. An additional challenge in surveillance is the difference in disease presentation, where Schistosoma co-infection with VL or CL can present as gastrointestinal disease (132, 133). Currently, it is not routine to screen for either of these parasites following gastrointestinal symptoms, so infections exhibiting these symptoms may go undiagnosed. In humans, CL lesions are smaller with faster healing times, and the immune response is predominantly characterized by an increase in a TH2 profile (127, 129). While mouse co-infection studies demonstrate predominant TH2 responses, TH1 responses are reduced, and lesion sizes, healing time, and parasitemia are increased (134, 135). This phenomenon has been discussed in a recent review (137). Perhaps, the mouse model does not adequately represent Schistosoma and Leishmania co-infection pathology in humans, and another model should be considered (e.g., hamsters). Nevertheless, mouse models remain an important tool since both humans and mice display strong TH2 responses following co-infection.

TRYPANOSOMA

Trypanosomiasis is often caused by one of two infecting species: Trypanosoma brucei responsible for African trypanosomiasis and T. cruzi responsible for American trypanosomiasis, both of which are also neglected tropical diseases (138).

African trypanosomiasis is primarily found in sub-Saharan Africa and is transmitted through the bite of a tsetse fly. Two main strains dominate infections of human African trypanosomiasis: T. b. gambiense and T. b. rhodesiense. These parasites infect the bloodstream and lymphatic system, and spread to the central nervous system, causing neurological symptoms. T. b. gambiense has been targeted by the WHO for total interruption of transmission by 2030 (139). Unfortunately, elimination is dependent on active case detection which is not feasible for T. b. rhodesiense due to its large animal reservoir and zoonotic nature (140).

American trypanosomiasis, or Chagas disease, is mostly found in Central and South America and is spread through the feces of reduviid bugs, also known as “kissing bugs.” The parasite enters the body through bite wounds or mucosal membranes and invades various tissues, notably the heart, and digestive and nervous systems. In chronic stages of Chagas disease, individuals may develop cardiac or gastrointestinal pathologies due to inflammation and tissue damage in response to the presence of T. cruzi nests, which, if left untreated, can be life-threatening (141).

Together, T. brucei and T. cruzi affect approximately 80 million individuals worldwide (142, 143), often in geographical locations where schistosomiasis is also present. Co-infections of these parasites have been reviewed previously (7), citing that in albino mouse models, Trypanosoma tends to suppress infection with S. mansoni/S. bovis (resulting in lower fecal egg counts per worm pair and reduced frequencies and diameters of egg granulomas) regardless of the order of infection. However, there are contrary data demonstrating co-infections present with increased splenomegaly and higher mortality than controls, with higher parasitemia of T. cruzi (144).

In correspondence with the latter data, a group from Brazil published work on T. cruzi infection 4 months post infection with S. mansoni (approximately 2 months into chronic infection) (145). This group found that when co-infected, animals experienced higher peak parasitemia as well as mean parasitemia compared to animals infected with only T. cruzi. In this study, co-infected animals presented reduced levels of IFNγ but higher levels of IL-10 compared to T. cruzi mono-infected animals. These findings suggest that S. mansoni infection may result in a reduced ability to mount a T. cruzi-protective TH1 response.

Furthermore, co-infected animals displayed a higher number of hepatic granulomas with marked confluence, higher cellularity, and lower fibrosis than animals infected with only S. mansoni, where granulomas were diffuse, well delimited, with low cellularity and higher collagen production. As granulomas mature, they become fibrotic to trap Schistosoma eggs and protect the host from hepatotoxins such as omega-1 (146, 147). This is especially important in the liver where eggs are not able to egress through the feces and are embedded in the tissue. Granulomas found in co-infected animals were more exudative-productive than they were fibrotic, which was inverse in animals which were only infected with schistosomiasis. This may show that when T. cruzi was present, there was a slowed progression of fibrosis around Schistosoma eggs, possibly resulting in a higher release of hepatotoxins.

The limited body of research surrounding trypanosomiasis and schistosomiasis co-infections focuses on T. cruzi and lacks information on T. brucei. Overall, the pathology resulting from both parasites together is controversial and differs in the literature. While early data demonstrate that co-infection with Trypanosoma may lead to protection from Schistosoma (148, 149), recent reports demonstrate that co-infection with the protozoan parasite may lead to increased liver pathology in schistosomiasis and increased Trypanosoma parasitemia (144, 145). One contribution to the discrepancies within results may be explained by the time at which animals were challenged which varied between papers. Trypanosoma-induced protection from Schistosoma may be temporal; protective effects seem to become weaker as the time between co-infections becomes longer (150). Furthermore, each study administered parasites with varying routes. Generally, 1,000–6,600 trypanosomes were administered intraperitoneally, and 25–40 schistosomes were given subcutaneously or percutaneously depending on the study; each of these variables can affect the data obtained. There is a lot of information yet to be gleaned from trypanosomiasis and schistosomiasis co-infection research. Since the data we have now come from animals, this leaves a large gap of knowledge in healthcare and epidemiological data. This underscores the need for further research in clinical settings to understand these co-infections within a human context.

TOXOPLASMA

Toxoplasmosis is caused by the obligate intracellular parasite Toxoplasma gondii, and although it can be found globally, it is more common in warmer, humid climates and in areas with poor sanitation (151). While only 10% of North Americans may have been exposed to T. gondii, in areas endemic for schistosomiasis and Europe, up to 60% of the population may have encountered this parasite (151). Although infection rates vary and European infections are often asymptomatic in immunocompetent individuals, toxoplasmosis presents with more severe manifestations in those who are pregnant and/or immunocompromised and those living in South America (152, 153). After acute infection, characterized by general flu-like symptoms, toxoplasmosis evolves into a chronic phase where parasites encyst into the brain and other organs in latent form. In cases of immunosuppression, these parasites can reactivate and result in pulmonary (154), cerebral (155), or ocular disease (156); infections in pregnant individuals may cause severe damage to the fetus, and congenital toxoplasmosis (vertical transmission) in children is likely to progress into clinical toxoplasmosis (155, 157).

One of the primary control cytokines expressed against T. gondii is IL-12, released upon parasite recognition by DCs, macrophages, and monocytes. In mice, this expansion is driven by parasite profilin recognition by toll-like receptors 11 and 12 (158, 159). While humoral immunity and antibody generating B-cells have been shown, in part, to mediate protection from toxoplasmosis (160, 161), type-1 immune effectors, such as IFNγ, are key contributors to parasite control and resistance (162).

Toxoplasma gondii is the most prevalent in Africa and parts of South America (163), and although its endemicity overlaps with Schistosoma in a broad range of locales, reports of co-infection in humans are indirect and limited to Tanzania (164, 165), Egypt (166, 167), and the Hunan province of China (168).

The earliest works of Schistosoma and Toxoplasma co-infection were conducted with S. mansoni in mouse models (169, 170) followed by the introduction of T. gondii. While effects were minimal when Toxoplasma preceded Schistosoma infections, both studies demonstrated that when mice were first infected with Schistosoma and then Toxoplasma, their mortality increased significantly. This was accompanied by increased levels of total IgE and TNFα, with a decrease in IFNγ and NO production. Recently, the increased severity of disease progression in co-infection was supported, again showing that mortality was increased when both parasitic infections were present (171).

Interestingly, small intestine pathology typical of Toxoplasma infection was ameliorated in mice that were first infected with Schistosoma. This includes histological changes such as inflammatory infiltration of the lamina propria, blunting of the villi, abnormal architecture, and areas of epithelial disruption. Of note, Schistosoma-co-infected animals displayed preserved goblet and Paneth cells, alongside a reduction of inflammatory cytokines. Ileal tissues also showed a decrease in expression of mRNA transcripts for IL-1β and nitric oxide synthase 2 and a decrease in activation of NFκB and p38 MAPK signaling (necessary for downstream cytokine production).

While T. gondii pathology was ameliorated in the intestines, Schistosoma co-infection became detrimental within the liver, which likely contributed to the increased mortality. Intracellular infection with Toxoplasma suppressed the S. mansoni-induced TH2 response necessary to prevent the release of Schistosoma egg hepatotoxins. Although visually clearer, with fewer Schistosoma-induced granulomatous formation, microscopic analysis showed that co-infected animals exhibited hepatic coagulative necrosis with extensive hepatocyte vacuolization, in support of previous work (169). Livers in these co-infected mice also showed fatty acid changes, with increased lipids and variations in metabolism (171).

Since IL-12 plays a significant role in toxoplasmosis control, one group explored its contributions to S. mansoni and Toxoplasma co-infections using IL-12 knockout mice (172). They found that IL-12-deficient mice did not share the same mortality as their wild-type counterparts when co-infected with both parasites. In fact, not only did they live longer, but splenocytes from these animals expressed lower levels of TNFα, IFNγ, NO, and increased (more than twofold) levels of IL-5, and IL-10. Further evidence that IL-12 contributes to the detriment of co-infected animals is that IL-12−/− animals did not display the same severe coagulative necrosis and cytoplasmic vacuolization seen in wild-type animals as the hepatic parenchyma and architecture were mostly preserved.

Together, the data suggest that the introduction of toxoplasmosis into an infection of schistosomiasis induces an immune shift away from typical TH2 responses, resulting in worsened schistosomiasis outcomes but ameliorated toxoplasmosis pathology in the small intestine (171). Knockout models of infection showed that IL-12 is a pertinent contributor to the pathology witnessed in co-infected animals, as in its absence animals lived longer and displayed a stronger TH2 profile (172). Still, the order of infection may play a significant role as studies demonstrate minimal changes when toxoplasmosis infection precedes schistosomiasis (169, 170). Like trypanosomiasis, to advance the field of toxoplasmosis-schistosomiasis co-infections, further clinical and epidemiological research is essential, with a focus on understanding the molecular mechanisms underlying these co-infections in human populations. This could inform better strategies for parasitic management and control in co-endemic areas.

INTESTINAL PROTOZOA

There are a variety of intestinal protozoa that infect humans causing infections ranging from asymptomatic colonization to severe disease. The most common pathogenic intestinal protozoa are Giardia duodenalis (syn. G. lamblia, G. intestinalis), Cryptosporidium spp. (typically C. hominis and C. parvum), and Entamoeba histolytica (173175). The classic manifestation of intestinal protozoa infection is diarrhea. Intestinal protozoa are spread worldwide, but typically, more infections occur in tropical climates and/or areas where access to sanitation is poor. Transmission occurs through the fecal-oral route, often involving consumption of contaminated food and water (176178). Both G. duodenalis and Cryptosporidium cause malabsorption and failure to thrive (179), and co-infections with the two protozoa are common (180). Many species of Entamoeba infect humans (176), but the majority are considered non-pathogenic. E. histolytica is primarily responsible for invasive disease, although only 10% of E. histolytica infections cause illness (181). Symptoms of amoebiasis include dysentery (sometimes severe) and extraintestinal symptoms, particularly amoebic liver abscess (182).

As all intestinal protozoa colonize the gut, there are some broad similarities in the immune responses they elicit. The intestinal epithelium and mucosa make up an important physical barrier, and breaching of this barrier is typically what leads to pathology. Gut mucosal immunity, such as IgA secretion, is important in intestinal protozoan immunity. Once the intestinal barrier is damaged by intestinal protozoan invasion, alarmins are secreted from intestinal epithelial cells, and innate immune cells are recruited to the site of damage. Cryptosporidium and E. histolytica infections drive a TH1-type response, with IFNγ having a protective role in both infections. G. duodenalis, in contrast, induces a TH17-skewed response, characterized by high levels of IL-17 (183, 184). IL-17 likely also plays a role in E. histolytica control; IL-4 and TNFα are associated with disease progression (185).

Intestinal protozoan infections occur worldwide, but as their incidence is increased in warm areas and locations with poor sanitation, there is significant overlap between them and schistosome infections. Co-infections of Schistosoma spp. and intestinal protozoa have been recorded in a variety of contexts (186190), but information on the immune response in co-infection remains limited. One prevalence study in North Samar, The Philippines, found that co-infections with S. japonicum and intestinal protozoa were highly prevalent. The co-infection rates with S. japonicum were 17.7% (for Cryptosporidium spp.), 14.6% (for G. duodenalis), and 9.9% (for E. histolytica). High rates of co-infection with a fourth intestinal protozoan, Blastocystis (44.9%), were also observed (189). There was a significant correlation between egg patent schistosomiasis and giardiasis in a community around Lake Albert in Uganda; however, specific rates of co-infection of Schistosoma spp. and G. duodenalis were not reported (188). Some studies outlined in a review found that co-infection with E. histolytica and Schistosoma spp. resulted in higher Entamoeba burdens and worsened morbidity in animal models and humans (7). A recent case report outlined the effects of co-infection with S. mansoni and E. histolytica in a 12-year-old boy from Ethiopia. After presenting to the hospital with bloody diarrhea, vomiting, weight loss, and fever, the patient was examined and diagnosed with ulcerative colitis and fulminant hepatitis due to his infections. The authors suggest that both infections contributed to the intestinal condition, but only S. mansoni to the hepatitis due to the lack of a liver abscess. The patient also showed signs of renal failure, but whether that was due to these infections was unclear. After treatment with PZQ and prednisone, the patient made a full recovery. This case highlights the need to better understand the pathology of intestinal protozoa and Schistosoma spp. co-infections, particularly in the context of bowel disease (190).

Unfortunately, more comprehensive research is lacking, and little to no information is available on the effects of co-infection with other intestinal protozoa on the immune response. It is important to better understand intestinal protozoa and schistosome co-infections as their pathologies and immune responses can be significantly altered by co-infection. Intestinal protozoa cause damage to the gut epithelium, which could have consequences for schistosome egg deposition and intestinal symptoms, given the proximity of where these pathogens reside. Likewise, the immune dampening seen in chronic schistosomiasis could influence the body’s ability to control intestinal protozoan infections. It is also important to consider the shared conditions that lead to schistosome and intestinal protozoan infections, namely, contamination of water with fecal matter and/or urine. WASH efforts will affect both schistosome and intestinal protozoan infection rates (191), and further integration of the multi-parasite landscape into control efforts will likely be beneficial for those at risk of infection.

MYCOBACTERIUM

Mycobacterium is a genus of bacteria comprising many species which cause disease in humans and animals. Arguably, the most prominent species of Mycobacterium is M. tuberculosis (Mtb). Mtb causes tuberculosis in humans and often affects the lungs as it is commonly spread through aerosols and respiratory droplets (192). Despite being a preventable and curable disease, TB infects 10 million people per year and kills 1.5 million (193).

About a quarter of the world has been infected with Mtb, though most will not develop disease, and some will even clear infection. Upon infection, Mtb may develop into active TB or latent TB (LTB), differentiated by the presence of symptoms and the ability to transmit infection to others. Although the immunological states corresponding to these various disease outcomes have yet to be elucidated, it has been shown that CD4+ T cell responses are important contributors of protection (194), especially those expressing type-1 effectors such as IFNγ (195).

This bacterium is present globally, though most individuals who develop severe disease live in low- to middle-income countries. According to the WHO, about half of all people with TB can be found in eight countries: Bangladesh, China, India, Indonesia, Nigeria, Pakistan, The Philippines, and South Africa (193). Of these countries, all except Bangladesh and Pakistan are endemic for schistosomiasis. Although S. mansoni has been shown to be an independent risk factor for active TB in Tanzania (196, 197), the broad impact of these infections when present together remains under-investigated.

For over 100 years, the bacille Calmette-Guérin (BCG) vaccine has been used to control Mtb (198). BCG is a live attenuated Mycobacterium bovis strain, and as such, BCG immunization and S. mansoni infection can be used as a model for Mycobacterium spp. and schistosomiasis co-infection. In mice infected with S. mansoni, it was found that there was an impaired control of BCG as colony-forming unit (cfu) counts were significantly higher in the lung, liver, and spleens of BCG immunized animals. Furthermore, splenocytes from these animals showed lower proliferation and expression of IFNγ than those which were solely immunized with BCG (199). When this model was extended to include Mtb infection, previous infection with S. mansoni decreased the protection offered by BCG. Not only did S. mansoni infection lead to increased cfu of Mtb in the lungs and liver, but it also resulted in a lower expression of both IFNγ and NO from restimulated spleen lymphocytes. This was accompanied by an increase in tissue damage, as lungs presented with increased cellular recruitment, less air spaces, and less organized granulomas which were diffuse without sharp demarcation from surrounding tissue (200). When Mtb lung granulomas are disorganized, B cell follicles are not well formed, and neutrophils lead to inflammation and damage (201), in contrast to Mtb lung granulomas containing distinct B cell follicles with T cells and macrophages, which are protective to surrounding tissue (202). While BCG is a useful tool to model Mtb infection, it has limitations since it is a different species, and it is attenuated. Still, the BCG data were supported years later in an animal model consisting solely of S. mansoni and Mtb. Monin et al. found that S. mansoni co-infection promoted lung fibrosis and inflammation in Mtb infection and altered the composition of Mtb granulomas (203). In co-infected animals, lung granulomas were found to be both type 2 (containing high arginase-1-expressing macrophages) and type 1 [containing inducible nitric oxide synthase (iNOS)-expressing macrophages] near each other, whereas animals infected with Mtb alone only exhibited type 1 granulomas. Arginase-1 (Arg-1) is a cytosolic enzyme, typically found in the liver, which hydrolyzes L-arginine during the urea cycle (204). In this context, Arg-1, from non-classically activated macrophages, competes with iNOS for L-arginine, limiting its ability to produce NO (205). Lungs from co-infected animals also were high in type 2 effectors, IL-4 and IL-13, matrix metalloproteases (MMP13), and neutrophil-produced myeloproteases. Interestingly, TB severity was reversed when co-infected animals were treated with an anthelmintic, suggesting that the presence of living adult worms contributes to an increased susceptibility to Mtb (203). In fact, when humans co-infected with helminths (Strongyloides, S. mansoni, and both) and LTB were treated with an anthelmintic, their restimulated PBMCs had lower frequencies of TREG cells and a rescue of Mtb-specific CD4+IFNγ+ T cells compared to before treatment (206).

Human co-infections of Mtb and schistosomiasis have recently been reviewed (37). Briefly, this systematic review demonstrated that S. mansoni impaired the immune response to Mtb and supported the hypothesis that infection with schistosomiasis can lead to a detriment of TB immunity, pushing LTB into reactivation (37). A study out of Switzerland showed that specifically S. mansoni, and not other helminths, was associated with clinical presentation among TB patients in Tanzania (196). Notably, when individuals were co-infected they were found to have less bacteria in their sputum, a finding which was also present in severely immunosuppressed HIV-positive individuals (207), inferring infection may be disseminated and extrapulmonary. This reduction in bacterial control could be attributed to immunomodulation from Schistosoma antigens. S. mansoni SEA has been shown to reduce the frequency of CD4+ T cells expressing IFNγ and TNFα from naïve human PBMCs stimulated with Mtb antigens 6-kDa early secretory antigenic target (ESAT-6) and 10-kDa culture filtrate protein (CFP-10) (208), perhaps as a result of schistosomiasis-dependent epigenetic IFNγ DNA methylation (209). Mtb-specific CD4+ T cell polyfunctional expression of TNFα, IL-2, and IFNγ has been associated with asymptomatic control of infection (210). When first exposed to SEA, CD4+ T cells stimulated with ESAT-6 and CFP-10 were less capable of doubly expressing IFNγ and TNFα, which made up 55.8% of the CD4+ T cell response from non-SEA-exposed cells (208). Schistosoma, and specifically SEA, are known inducers of TH2 immune responses which have been shown to impair TB immunity (211, 212).

However, when naïve human PBMCs (213) and monocyte-derived macrophages (214) were primed with SEA in vitro, they exhibited improved control of intracellular Mtb. Furthermore, Mtb-specific type 1 CD4+ T cells were maintained in a Kenyan cohort of S. mansoni- and Mtb-co-infected individuals. Curiously, these cells were found to be TH2 lineage cells (CCR4+) capable of expressing TH1 cytokines IFNγ, TNFα, and IL-2 (215), although further research is necessary to determine whether these polyfunctional CD4+ T cells were a by-product of schistosomiasis TH2 immunity. Separately, the role of monocytes (216) and TGFβ-producing TREG cells (217) in S. mansoni and active TB co-infection have also been investigated. Despite these data, it is still unclear why TB outcomes are worsened in schistosomiasis-co-infected individuals.

Conversely, there are limited data describing the influence of Mtb on outcomes of schistosomiasis, despite the bacterium’s ability to alter immune responses to Schistosoma antigens. PBMCs were drawn from Kenyan individuals and stimulated with S. mansoni soluble worm antigen preparation (SWAP). T cell cytokine production to SWAP was found to be dominated by γδ T cell-expressed IFNγ and TNFα. When cases of active TB were assessed, there was a significant decrease in the amount of responding γδ T cells and an increase in their expression of IL-4 (218). While interesting, it has yet to be elucidated what effect these immune changes may have on schistosomiasis.

Although the immunological effects of schistosomiasis on tuberculosis are unclear, the consensus suggests that helminth-induced TH2 responses impair TH1 effectors necessary for Mtb control as summarized in Fig. 4. Schistosomiasis may even increase TREG responses which, with TH2 immunity, hinder IFNγ and TH1-mediated protection from TB. Mtb, on the other hand, has demonstrated the ability to dampen γδ T cell responses to Schistosoma antigens (216, 218). Although most of the literature around Mycobacterium and Schistosoma is composed of Mtb research, recent publications have exposed the importance of schistosomiasis on Mycobacterium leprae, supporting the hypothesis that helminth infections may influence the transmission and risk of active leprosy (219221). More work is needed to elucidate whether the same mechanisms between schistosomiasis and Mtb could be affecting the pathobiology of M. leprae.

Fig 4.

Fig 4

Proposed immunological effects of schistosomiasis and tuberculosis co-infection. In co-infections of schistosomiasis and tuberculosis, the helminth promotes type 2 responses and type 2 granuloma formation, IL-4 expression, and TREG responses which may lead to worsened TB outcomes. Research suggests that schistosomiasis reduces CD4+ T cell IFNγ responses crucial for tuberculosis immunity, while tuberculosis reduces helminth-specific γδ T cell responses.

SALMONELLA

Transmitted through the ingestion of contaminated food or water, infections of Salmonella can be found globally. Salmonella is responsible for 180 million cases of diarrheal illnesses that occur yearly, representing 9% of total diarrheal cases. Despite this number being lower than the etiologies of diarrheal disease tracing back to other pathogens (e.g., norovirus, enterotoxigenic Escherichia coli, Shigella spp., and Giardia spp.), a disproportionate number of deaths can be attributed to Salmonella (41% of all diarrheal disease-associated deaths) (222). This genus comprises various species with diverse degrees of severity but is best known for S. enterica subtypes Typhi and Paratyphi causing typhoid and paratyphoid fevers, respectively (223), as well as invasive non-typhoidal S. Typhimurium (iNTS) (224). These species are not evenly distributed around the globe as iNTS is found in Africa, while typhoidal serotypes (Typhi and Paratyphi A) are more commonly found in Southeast Asia (222); both of which are regions endemic for schistosomiasis.

Salmonella are rod-shaped gram-negative bacteria, and despite their ability to live extracellularly, most associated pathology comes from their intracellular life cycle (225). Most manifestations of disease are self-limiting; however, severity depends on serotype involved and the health status of the host. In general, children, the elderly, and immunocompromised individuals are more at risk of infection with Salmonella than healthy individuals (226). In human infections, Salmonella typically presents in four manifestations: enteric fever, gastroenteritis, bacteremia, and chronic carrier state (227).

S. enterica possesses several virulence factors which cause the bacterium to not only activate the immune system but allow it to resist and evade it. Using innate immune cells as a reservoir, Salmonella spp. are capable of invading macrophages and growing and persisting within them. In addition to prevention by vaccination (228), salmonellosis can be treated with oral fluids and antibiotics for those who are immunocompromised (229). Keys of protection from Salmonella include humoral and cellular immunity (230), some cytokines including IL-6, IL-17 (231), and IFNγ (232), as well as bacterial control via the microbiome (233).

The pathogenicity of Salmonella during infections of schistosomiasis has been reviewed recently within the context of the microbiota (234). During cases of co-infection, disease manifests as prolonged fever and hepatosplenomegaly, alongside chronic septicemic salmonellosis (235). Human reports of schistosomiasis and Salmonella co-infections have been seen in Egypt (236, 237), Ethiopia (238), Gabon (239), the Democratic Republic of Congo (240), and Tanzania (241). As early as 1978, it was shown that patients experiencing co-infections of schistosomiasis and Salmonella presented with renal problems (237), and others with prolonged bacterial infection exacerbated pre-existing schistosomal glomerulopathy (242, 243). Interestingly, due to schistosomiasis worms harboring Salmonella bacteria within their intestines and tegument (234, 244, 245), Salmonella-targeting antibiotics became less effective (246). Patients often did not respond to antibiotic treatment, presenting with recurrent fever, until anthelmintics were administered (235, 239, 246).

Multiple studies have demonstrated enhanced colonization of Salmonella spp. in animals infected with Schistosoma spp. (235, 247249). This effect could be seen in early mouse work where macrophages from S. mansoni-infected mice were less capable of killing S. Typhimurium (250, 251). When mice were injected with S. mansoni eggs, S. Typhimurium led to increased inflammation in the gut lumen (248), and in mice chronically infected with S. mansoni, S. Typhimurium co-infection worsened liver injuries (increased expression of inflammasome and apoptotic proteins) and shifted egg responses from protective granulomas to congested and necrotic (249). This co-infection altered the immune landscape within mice from profibrogenic TH2 responses to anti-fibrotic TH1 responses, paralleled with a decrease in IL-4 and IL-5, and increased IL-10, IFNγ, and IL-2. Co-infected animals not only exhibited compounded liver pathology but also increased bacterial loads (248). Additionally, hamster studies showed that S. mansoni and S. Paratyphi A co-infection led to increased mortality (252). Curiously, despite this exacerbation of salmonellosis, mice co-infected with intestinal schistosomiasis exhibited lower worm and egg burdens. S. japonicum and S. Typhimurium co-infection resulted in lower burdens of Schistosoma worms and eggs, than S. japonicum mono-infection. This was accompanied by an increased expression of IFNγ and a decreased expression of IL-4 in the serum showing systemic shifts in immune response (253). Even in a murine co-infection model of S. mansoni and S. Typhimurium, it was observed that co-infected mice exhibited a significant reduction in hepatic eggs compared to those infected with S. mansoni alone (249).

While touched on here, Salmonella and Schistosoma co-infections are complex and understudied, occurring primarily in regions of Africa. Together, these infections lead to prolonged fever, hepatosplenomegaly (235), and unique challenges in treatment due to Salmonella habitation within schistosomiasis worms (234, 244, 245). Although research suggests altered immune responses and reduced Schistosoma parasite burden in co-infected animals, mortality levels and salmonellosis pathology increased. Further studies are needed to elucidate the mechanisms, clinical impact, and potential therapeutic strategies for these co-infections in human populations.

URINARY TRACT INFECTION-CAUSING AGENTS

Urinary tract infections (UTIs) are commonplace worldwide. In fact, more than 50% of all females will experience a UTI at some point in their lives (254). Despite the predisposition of young females to UTIs, there is a significant population of males who experience UTIs (254); UTI prevalence in elderly males and females is similar (255). A myriad of pathogens (mostly bacterial, however, also protozoan and fungal) can cause UTIs, resulting in benign illnesses to serious complications such as sepsis and re-current infection (254).

Due to their global prevalence, UTIs can easily be found in locations endemic for schistosomiasis (256) and have been reported as co-infections in Cameroon (257), Ghana (48), Niger (258), and Nigeria (259), to name a few. Since symptoms of schistosomiasis and UTIs are similar, clinicians sometimes misdiagnose and mistreat infected individuals (260, 261). Most reported cases of dual infection involve S. haematobium, or urinary schistosomiasis; however, cases of S. mansoni and UTIs have also been described (262266).

Interestingly, while females are more commonly infected with UTIs, field data demonstrate that males are more often infected with S. haematobium (267269). When both UTIs and schistosomiasis data were compared together, it was found that co-infection in Nigeria was age and gender dependent, with a higher incidence observed in young males (270). Furthermore, most groups conclude that S. haematobium (259, 262, 265, 271277) and S. mansoni (264, 265, 276) infections increased the incidence of concomitant UTIs. Two groups declared no significant association between S. haematobium and concurrent infection with urinary pathogens (258, 278), though it was found that co-infections with both significantly increased from 0.42% to 7.712% when moving from the dry to rainy season (278).

In schistosomiasis-positive individuals, the most common pathogen infecting the urinary tract was Escherichia coli, though groups also found significant amounts of Klebsiella spp., Staphylococcus aureus, Staphylococcus saprophyticus, Proteus vulgarius, Pseudomonas aeruginosa, Streptococcus spp., and Candida albicans (262, 267, 269, 272, 279) in urine specimens. Trichomonas vaginalis (269, 280, 281), Neisseria gonorrhoeae (281), and Salmonella Paratyphi A (282) have also been reported to infect the urinary tract of individuals with schistosomiasis.

Some have postulated that infection with schistosomiasis poses a risk factor for UTIs due to egg spines causing damage to urinary tissues, providing bacterial entry sites and blood which acts as a bacterial-culturing medium (283, 284). Animal studies using models of co-infection demonstrate that S. haematobium eggs increase susceptibility to uropathogenic E. coli bacteriuria in BALB/c mice due to an increased expression of IL-4 (285) and a reduction of invariant natural killer (NK) T cell-mediated bacterial clearing (286). Additionally, administration of S. haematobium IPSE (an egg-derived protein), despite showing no changes in bacterial cfu, reduced anti-microbial peptides in the bladder and may work in concert with IL-4 to promote the establishment of bacterial infection (287).

UTIs are a widespread issue co-occurring often with schistosomiasis in many African countries. These co-infections are challenging to diagnose due to their similarity in symptom manifestation (260, 261). S. haematobium and S. mansoni are linked to increased UTI risk (259, 262, 264, 265, 270277), potentially due to parasite damage to host tissue and parasite impact on the immune system, especially in young males (270). Preclinical studies support this hypothesis, showing that schistosomiasis can make animals more susceptible to uropathogenic E. coli infections (285, 286). Given S. haematobium is associated with genital schistosomiasis (4850), and the urinary tract and genitals are in proximity, UTI co-infections within this context should also be considered. Further research would help our understanding of the mechanisms by which these infections interplay to develop effective strategies for diagnosis and treatment in co-endemic regions.

SEVERE ACUTE RESPIRATORY SYNDROME CORONAVIRUS 2

Unlike most bacteria, viruses cannot replicate outside of living cells; the survival of a virus in nature depends on maintaining serial infections (i.e., a chain of transmission; causing disease is neither required nor necessarily beneficial) (288). Still, viral infections can have devastating impacts on the lives and livelihoods of individuals around the globe; for example, coronavirus disease 2019 (COVID-19) alone has led to more than 6.9 million deaths to date (289, 290).

COVID-19 is a respiratory disease that has had devastating impacts worldwide since its declaration as a pandemic by the WHO and has been described as the worst epidemic of the century (291, 292). With over 500 million confirmed COVID-19 cases, the causative virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has a rapidly expanding genealogy that now warrants classification of at least 13 variants. Moreover, this virus appears to be becoming endemic, with mutations in the N-terminus and receptor-binding region, including p.Glu484Lys which can be found in the most dangerous variants (293). Variants of concern include Alpha, Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529), with Delta and Omicron being the most severe (294). Since the start of the outbreak, many studies have shown a dramatic increase in COVID-19 in low- to middle-income countries compared to developed countries with better infrastructures and health policies (295). Other factors and various socioeconomic indicators, including gross domestic product per capita, human development index, and differences in the timing of disease onset, have been used to explain geographical differences in the burden of COVID-19 (296, 297).

Neutralizing antibodies have typically been the gold standard of efficacy for viral clearance; however, evidence is growing that CD8+ T cell responses and type-1 immunity are important for SARS-CoV-2 viral clearance, especially in breakthrough infections (298300). Thus, the immunomodulatory effects caused by schistosomes are a double-edged sword as they may protect the host from the detrimental inflammatory consequences of COVID-19 or support viral infection (301).

Co-infections of schistosomiasis and SARS-CoV-2 appear unlikely to be critical factors in determining the severity of the disease (302), albeit it is possible. Based on an observational cohort study (303), it was shown that parasites which induce TH2 responses resulted in a reduced risk of severe COVID-19 by dampening immune hyperactivation which is linked with pathogenesis of severe viral disease (304). When stratified by species, S. mansoni was among the parasites that implied a lower probability of severe COVID-19. Although schistosomiasis was associated with decreased SARS-CoV-2 severity, it is important to consider other helminths co-endemic in these regions, such as STHs, which may also be responsible for improving viral disease outcomes (305).

Despite the lack of information on the direct effects of COVID-19 on schistosomiasis, the pandemic has severely affected schistosomiasis transmission indirectly. With social distancing and the halting of PZQ mass drug administration, Brazil has seen a reduced amount of the population tested for schistosomiasis and, therefore, underestimated positivity rates. Furthermore, treatment of positive cases was only approximately 77% (306), inferring the potential for increased transmission of infection in the country. Moreover, in China, schistosomiasis is caused by S. japonicum and is spread by the snail vector Oncomelania hupensis. COVID-19 closures allowed this snail species to return to its natural habitat in Wuhan, increasing the risk of schistosomiasis spread (307).

Mathematical modeling has been used to determine the effects of COVID-19-caused postponement of MDA and other mitigation strategies on elimination plans for S. haematobium and S. mansoni. For both species, elimination plans have been estimated to be delayed by up to 2 years in moderate- and high-transmission areas (308). To reduce schistosomiasis infections and to attempt to achieve WHO elimination goals, these mitigation strategies need to be reinstated and maintained through future healthcare emergencies.

It is currently unclear the effects COVID-19 and schistosomiasis have on each other. Aside from the possibility of helminth-induced TH2 responses ameliorating immune hyperactivation in severe viral disease and the contributions of the global pandemic to schistosomiasis transmission, the effects of this co-infection are controversial. SARS-CoV-2 is a relatively newly emerged pathogen, and as data are collected, these associations may become clearer. Future studies are necessary to confirm how these pathogens interact and how this interaction affects the host.

HUMAN IMMUNODEFICIENCY VIRUS

Human immunodeficiency virus causes acquired immunodeficiency syndrome and is one of the world’s most serious public health challenges. There were approximately 38.4 million people across the globe living with HIV in 2021 (309). Of these, 36.7 million were adults, and 1.7 million were children. An estimated 1.5 million individuals worldwide acquired HIV in 2021 (310), demonstrating consistent transmission. Currently, there is no cure for HIV/AIDS; however, many medications can control HIV and prevent complications (309). These medications are called antiretroviral therapy (ART). ART therapy aims to lower the amount of HIV in the blood to undetectable/untransmissible levels. HIV infection is one of the main causes of morbidity and mortality worldwide, with most of the disease concentrated in sub-Saharan Africa (311).

HIV primarily targets CD4+ T cells. Following transmission, HIV establishes itself in the mucosal tissues and spreads to the lymphoid organs within days. The virus becomes detectable in the blood around day 10 and continues to spread exponentially over the next few weeks, though in some cases, seropositivity can take up to 3 months. Most viral shedding takes place at this point (312). The immune system, with substantial contributions from CD8+ T cells, is able to achieve some control over HIV replication (313). This leads to the establishment of a “set point” where HIV replication remains relatively stable, often lasting for years (312). HIV causes a progressive loss of CD4+ T cells and a variety of immunological abnormalities through mechanisms that are likely to be multi-dimensional and are not yet fully understood (314). Pronounced immunodeficiency and development of characteristic infectious or oncologic complications occur after several years (312).

The HIV pandemic is generalized across many regions of sub-Saharan Africa, and its geographical overlap with schistosomiasis is concerning (315). In 2020, the Joint United Nations Programme on HIV/AIDS produced maps of Malawi depicting HIV incidence among young females (aged 15–24 years) at the district level for the year 2019 and of S. haematobium prevalence in the school population for the years 2012 and 2013. These maps suggest that in districts along Lake Malawi, there may be an ecological link between schistosomiasis and HIV incidence in young females (36).

Schistosomiasis appears to be a cofactor in the spread and progression of HIV/AIDS in areas where both diseases are endemic; increased emphasis on the treatment of schistosome infections in persons at risk of HIV/AIDS should be pursued (316). A further study suggests that people with chronic schistosomiasis may be more likely to become infected with HIV type-1 (HIV-1) and be impaired in viral control once infected (317). Highly active antiretroviral therapy (HAART) can effectively control viral replication in the blood but may be less effective in the gut where the majority of CD4+ T cells reside. Extensive loss of CD4+ T cells in the gut during acute HIV-1 infection suggests that, in addition to HAART, it is important to prevent viral infection of susceptible cells in the gut after initial exposure (318).

Immunological changes occurring during schistosomiasis may increase HIV transmission risk (36). It has been shown that schistosomiasis-induced pathological changes (such as inflammation and damage to the mucosal lining of genital and rectal areas) can increase the shedding of HIV and facilitate its transmission during sexual contact (316). Therefore, PZQ treatment may be beneficial in preventing HIV susceptibility (319, 320). In the northern hemisphere, men who have sex with men have been disproportionately affected by HIV-1 (321); however, it is unclear how this population compares in areas endemic for schistosomiasis. Several studies show that schistosomiasis, specifically female genital schistosomiasis among adolescents and young adults, may be associated with a higher risk of HIV acquisition (36, 320). Female genital schistosomiasis is primarily caused by S. haematobium, and less commonly by S. mansoni, and affects an estimated 20–56 million young females (322). Co-infections of HIV and female genital schistosomiasis cause mucosal changes in the intravaginal epithelium of the cervix, fornices, and vagina, as well as infertility when the upper genital tract is involved (323). In addition to Schistosoma-induced immunological changes, mast cells have been described as a reservoir of HIV-1 that can be induced by schistosome infections (324). Though it has been demonstrated that schistosomiasis may increase the risk of HIV-1, it is unclear what effect HIV-1 infection has on susceptibility to schistosomiasis (315).

Schistosomiasis may also alter immune responses to HIV. Immunoregulatory responses associated with helminth infection downregulate TH1 immune responses associated with viral control (325). In the TH2 cells associated with helminth infection, HIV replicates more easily (326, 327). Likewise, activated immune cells, as found around Schistosoma eggs in genital lesions or in neighboring sites, are predisposed to HIV infection (326, 328, 329). Schistosoma eggs elicit a complex cellular and humoral immune response, including upregulation of cellular CD4 and chemokine co-receptors, used by HIV-1 to enter host cells. This further increases the risk for those people with female genital schistosomiasis of being infected and transmitting HIV (49). Despite the increased risk of HIV-1 acquisition in schistosomiasis-infected individuals, a study from Mwanza, Tanzania showed that viral loads in adults were not elevated, and HIV progression was not worse in those co-infected (330, 331).

It has been found that individuals co-infected with HIV-1 and S. japonicum exhibited a lower frequency of CD4+ T cells and a higher frequency of CD8+ T cells (332). Individuals co-infected with HIV-1 and S. mansoni displayed an extra enlarged left hepatic lobe which suggests that co-infection may be associated with severe hepatosplenic disease (333). Other liver ailments have also been attributed to HIV-1 and schistosomiasis, including periportal fibrosis (334), and higher hepatotoxicity in co-infected patients on ART (335). Interestingly, in sub-Saharan Africa, it has been shown that HIV-1 infection reduced egg excretion in individuals infected with S. mansoni (315, 336, 337), suggesting that Schistosoma diagnosis by microscopy may not be ideal in co-endemic regions. The co-occurrence of HIV and schistosomiasis may be a larger hindrance to the elimination of these pathogens than initially suspected. As many populations lack accessible MDA of PZQ and ART, Schistosoma/HIV co-infections may be synergizing to increase pathology and HIV transmission to uninfected individuals.

Co-infection with HIV and schistosomiasis presents a significant public health challenge. Schistosomiasis may increase susceptibility to HIV (36, 317) and alter immune responses (36), particularly in female genital schistosomiasis (36, 320), potentially increasing HIV transmission (Fig. 5). This is especially dire in regions like sub-Saharan Africa where an estimated 30% of HIV+ individuals were not on antiretroviral treatment at the end of 2019 (338). Furthermore, the effects of HIV on schistosomiasis are noteworthy, as co-infected individuals may experience severe hepatosplenic disease (333). In adults, schistosomiasis typically precedes HIV-1 infection. In children who are born HIV+, however, HIV-1 infection precedes schistosomiasis which may present an altered immunological picture. The complex interactions between this virus and this parasite require further investigation. Additionally, their co-endemicity may hinder elimination efforts for both infectious diseases, particularly in regions with limited access to preventative measures and treatment.

Fig 5.

Fig 5

Proposed immunological effects of schistosomiasis and human immunodeficiency virus co-infection. Schistosomiasis exacerbates TH2 responses, hindering TH1 responses, including CD8+ T cells vital for viral control. Additionally, schistosomiasis [especially female genital schistosomiasis (FGS)] induces immune and pathology changes that may increase HIV susceptibility and transmission risk. Finally, schistosomiasis causes CD4+ T cell and mast cell expansion prior to HIV infection, leading to a larger HIV reservoir in the host. Post-infection, HIV contributes to a decline in CD4+ T cell counts. Co-infections of schistosomiasis and HIV may lead to increased hepatosplenic disease.

HEPATITIS VIRUSES

Hepatitis viruses are a group of viruses that primarily affect the liver, causing inflammation and damage to the organ. Acute viral hepatitis is caused by several different hepatitis viruses, including hepatitis A (HAV), hepatitis B (HBV), hepatitis C (HCV), hepatitis D (HDV), and hepatitis E (HEV) (339). These viruses have different characteristics, routes of transmission, and potential health effects. HAV and HEV can be found worldwide, though they are more prevalent in areas with poor sanitation and hygiene practices. These viruses are primarily spread by fecal-oral transmission, resulting in both sporadic infections and large-scale outbreaks due to contaminated food or water (340, 341). The incidence of HBV varies by geography, with significantly higher rates in sub-Saharan Africa, parts of Asia, the Middle East, and the Amazon (342). HCV has a global distribution, affecting people in all regions of the world. Egypt, parts of central and eastern Asia, and some eastern European countries are areas of significant HCV prevalence (343). HDV is a less common hepatitis, and it requires co-infection with HBV to cause illness (344). Treatment for hepatitis viruses varies depending on the type of hepatitis and the underlying cause. With supportive care, such as rest and proper nutrition, hepatitis A and E infections are often self-limiting, and most people recover on their own within a few months. However, hepatitis B and C can lead to chronic infection and long-term liver pathology if left untreated. Treating chronic HBV and HCV aims to control the virus, reduce liver damage, and prevent complications (344). Nucleoside or nucleotide analogs such as entecavir, tenofovir, lamivudine, and adefovir are oral medications that directly target the virus and inhibit its replication. The choice of medication depends on several factors, including the severity of liver disease, viral load, and the presence of certain viral mutations (345). Regular monitoring of liver function and viral load is important to assess treatment response. Direct-acting antivirals (DAA), such as sofosbuvir, ledipasvir, velpatasvir, glecaprevir, and pibrentasvir, are now the standard of care for HCV. These medications specifically target the virus and have a high cure rate (346). DAA regimens vary depending on the specific genotype of the virus and may involve a combination of different medications taken orally for a specific duration, usually from 8 to 12 weeks (347).

Depending on the specific hepatitis virus involved, immune responses also differ. Infection with a hepatitis virus results in activation of the innate immune system as the first line of defense. Pro-inflammatory interferons contribute to the inhibition of viral replication (348, 349). NK cells and macrophages are also involved in the elimination of infected cells. CD4+ helper T cells play a critical role in coordinating the immune response, while CD8+ cytotoxic T cells directly target and kill infected cells, limiting the spread of the virus (350, 351). B cells are responsible for the production of antibodies that can neutralize the virus and prevent its entry into cells (352). In some cases, the immune response alone is not sufficient to eliminate hepatitis infections; this is particularly true for HBV and HCV. Chronic hepatitis infections can cause persistent inflammation and liver damage (348, 353).

Schistosomiasis and hepatitis co-infection is reported in many countries, notably in Egypt (354). Here, the prevalence of schistosomiasis in patients with HCV infection was reported to be between 27.3% and 50% across two studies (355, 356). Furthermore, a high prevalence of HCV and S. mansoni co-infection was reported among workers employed in sewage treatment plants (354). Co-infection of HBV and S. mansoni ranged from 19.6% to 33.0% (357). It is interesting to note that a high prevalence was also demonstrated in China, where about 58.4% of the patients with chronic S. japonicum had HBV (354) and in Hubei, specifically, where the prevalence of HBV in schistosomiasis patients was much higher (25.37%) compared to those without schistosomiasis (0.62%) (358). The risk of becoming infected with HBV was significantly higher in patients with HCV and schistosomiasis compared to those without schistosomiasis. Similarly, in Egypt, the prevalence of HBV was higher in patients already co-infected with HCV and schistosomiasis (12.8%) than in those without schistosomiasis (8.5%) (356). Moreover, chronic schistosomiasis patients have had a higher risk of HCV/HBV co-infection due to parenteral schistosomiasis treatment using non-sterile syringes, blood transfusion for anemia, surgery, and endoscopy (359, 360).

One review has examined the pathology of Schistosoma and HBV/HCV co-infections between 1975 and the end of 2014 (361). The authors found that the clinical course of illness once an individual was co-infected with Schistosoma and either HBV or HCV was typically more severe than those which were mono-infected. For both viruses, co-infection resulted in detrimental pathologies leading to higher mortality. Other reviews have also demonstrated the importance of hepatitis-schistosomiasis co-infections (357, 362, 363). One postulated that vaccination may be a solution to co-endemicity (364), though a recent preprint demonstrated that Schistosoma infection blunts HBV-vaccine responses, highlighting that it may be necessary to clear the helminth infection pre-vaccination (365).

The effects of schistosomes on HBV and HCV have been thoroughly examined by Bullington et al. (366). In patients with hepatosplenic schistosomiasis, HBV and HCV co-infections have been shown to be associated with a marked depression of cell-mediated immune responses (364).

In cases of HCV, some studies reported impairment of HCV-specific CD4+ T cell responses. Co-infections of hepatitis C and schistosomiasis also resulted in a promotion of TH2 responses (IL-4, IL-10) over TH1 responses, which may explain the synergistic relationship of Schistosoma-HCV on both liver fibrosis and mortality (354). In co-infected patients, increased HCV RNA titers, histologic activity, and incidence of cirrhosis/hepatocellular carcinoma were observed (367), in addition to liver and spleen stiffness (368).

Although the effects of Schistosoma on HBV infection remain a subject of debate as schistosome-induced factors like IFNγ and NO can inhibit HBV replication (369) and one study did not find a clear association between schistosomiasis and worsening of HBV infection (370), most reviews conclude that HBV and schistosomiasis co-infections also led to poorer prognoses (354, 361). Chronic schistosomiasis and HBV co-infection may progress from advanced cirrhosis to hepatocellular carcinoma, with co-infection found to exaggerate liver function abnormalities, histological changes, and fibrosis more than HBV mono-infection (371373). This was supported by a study which reported that HBV in association with hepatosplenic schistosomiasis led to worse outcomes (358).

In conclusion, hepatitis viruses, with their various types and routes of transmission, present distinct challenges in terms of diagnosis, treatment, and the immune responses they elicit. Co-infection with schistosomiasis further complicates clinical course and outcomes, often resulting in more severe liver pathologies and increased mortality (354, 361). As we navigate this complicated landscape, primary prevention of HCV and HBV infections remains crucial, especially in regions where schistosomiasis is endemic. There is a pressing need for continued research to fully understand the dynamics of these co-infections. This research should encompass various aspects, including the potential influence of co-infections on mass HBV vaccination, which will aid in developing more effective strategies for managing and preventing these infections.

OTHER PATHOGENS AND MULTIPLE CO-INFECTIONS

Cytomegalovirus (CMV) is a common virus belonging to the herpesvirus family (374). CMV is common worldwide infecting a substantial number of individuals at some point in their lives, though its prevalence varies in different population groups and geographical regions (375). CMV seroprevalence is generally higher in women, older age groups, lower socioeconomic status, and low- to middle-income countries (376). Among women of reproductive age, global CMV seroprevalence ranges from 45% to 100% (376). CMV can be found in various bodily fluids, including saliva, urine, blood, semen, and breast milk. Common routes of transmission include sexual contact, organ transplantation, blood transfusion, and vertical transmission during pregnancy (377). Vertical transmission involves CMV passing from a pregnant person to their fetus, resulting in congenital CMV infection (378). Congenital CMV infection can cause a range of health problems including hearing loss, developmental delay, and neurological abnormalities (379).

CMV is of particular concern for people with weakened immune systems, such as those with HIV/AIDS, organ transplant recipients, or those undergoing chemotherapy. The immune response to cytomegalovirus is complex and involves several different components of the immune system (380382). NK cells play a crucial role in recognizing and eliminating CMV-infected cells (383).

Since NK cells are important for CMV elimination and hepatic NK cell function is inhibited via the T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain receptor in schistosomiasis-induced liver fibrosis, co-infections of CMV and schistosomiasis may potentiate viral load (384, 385). Also, it is worth noting that individuals with weakened immune systems, such as those with advanced schistosomiasis or other chronic illnesses, may be more susceptible to CMV infection and its associated complications which highlights the importance of accurate diagnosis and the appropriate management of these conditions.

Differently, arboviruses, or arthropod-transmitted viruses, are a group of viruses that are transmitted to humans and other animals primarily through the bites of infected arthropod vectors such as mosquitoes, ticks, and sandflies (386). Arboviruses are found in different parts of the world, and their distribution is influenced by the presence of suitable vectors and reservoir hosts (387). The presence and abundance of arthropod vectors strongly influence the transmission dynamics of arboviruses (388). Factors such as vector species, their distribution, behavior, and vector competence contribute to the epidemiology of arboviral diseases (388). Some arboviruses have reservoir hosts, typically mammals, which maintain the virus in nature and can serve as sources for transmission to vectors (389). Infected individuals or vectors can carry the virus to regions where it is not endemic, potentially leading to localized outbreaks or establishing new transmission cycles (390). Climate factors, including temperature, precipitation patterns, and habitat changes, can influence the distribution and abundance of arthropod vectors and alter the epidemiology of arboviral diseases (391). Climate change may even expand or shift the geographic range of certain vectors and increase the risk of arboviral transmission in previously unaffected areas (392). Notably, arboviruses include a diverse group of viruses, such as dengue, Zika, chikungunya, yellow fever, and others, each of which has different characteristics and interactions with the host’s immune system (393).

There is limited research specifically on the association between schistosomiasis and arboviruses; schistosomiasis primarily affects human blood vessels and can lead to chronic inflammation, immune modulation, and alterations in the immune response (394), and the susceptibility and severity of arboviral infections may be influenced by this altered immune environment (395). Some studies suggest that co-infection or previous exposure to schistosomiasis may be a modulator of the immune response to arboviral infections (396), which can have an impact on the clinical outcome or severity of the disease (397). Despite the lack of research on these co-infections, these pathogens are often co-endemic; for example, in Brazil, where a young person infected with Zika virus exhibited testicular inflammation and extensive loss of testicular structure, with granulomas induced by S. mansoni eggs (398). In areas where schistosomiasis and arboviruses co-occur, it is important to address both infections through a comprehensive set of public health interventions.

Although extensive, this review is not exhaustive. Schistosoma spp. are prevalent in 78 countries worldwide, resulting in chronic infections, making schistosomiasis a common co-infecting pathogen. Some examples of other Schistosoma co-infecting pathogens which were not discussed in this review include Helicobacter pylori (399), human papilloma virus (51), and human T lymphocyte virus-1 (400) to name a few. Moreover, it is important to note that this review did not address potential co-occurrence of fungal infections alongside schistosomiasis.

Adding more depth and convolution to Schistosoma co-infections are individuals infected with three (or more) pathogens. One example recently published is of a Polish traveler who returned from the Democratic Republic of Congo, presenting with disseminated skin rash and eosinophilia, and was diagnosed with Schistosoma sp., Strongyloides stercoralis, Trichuris trichuria, and Blastocystis sp. (401). Another example is a Ghanaian immigrant who was seropositive for Schistosoma sp., filaria, and Strongyloides sp. (402). Some epidemiologic studies have been completed in cohorts of people with Schistosoma co-infections with two other pathogens. Cases of anemia have been seen in Ethiopian children infected with Schistosoma mansoni, Plasmodium sp., and hookworm (100). Schistosoma mansoni was also reported to increase the risk of TB disease in HIV+ Kenyan adults (403), while it did not contribute to mortality or recurrence of non-typhoidal Salmonella bacteremia in HIV+ Malawian adults (404).

Given the infinite potential for co-infections involving Schistosoma sp., it is unfeasible to comprehensively address all of them within a single review. However, it is evident that helminthic parasites, by modulating the immune system, play a significant role in influencing other infections and diseases.

CONCLUSIONS

Despite their prevalence, limited work has been published on the co-infections of Schistosoma spp. and other pathogens. Unfortunately, this comes with the territory of many neglected tropical diseases. Yet, the urgent need to obtain insight into the epidemiology, pathogenesis, and driving immune mechanisms of Schistosoma co-infections can be supported by the sheer number of individuals at risk of disease (8). Figure 6 depicts a summarized view of the prevalence of co-infecting pathogens with Schistosoma spp. As this map is broadly representative and cannot display whole special regions, we can appreciate the underestimation of co-endemicity between pathogens.

Fig 6.

Fig 6

Schistosoma and co-infections world map. The navy-blue shading on the map indicates regions where Schistosoma spp. are endemic. Within these areas, colored dots mark co-endemic regions for other pathogens. Geographical regions are categorized as South America, Africa, the Middle East, India, China, and the Southeast Asian nations. Intestinal protozoa were omitted as they are ubiquitous. Salmonella, SARS-CoV-2, and cytomegalovirus are also ubiquitous and have been marked in areas where they are most prevalent.

Being a chronic infection, which has evolved with humans (405), schistosomes carefully balance parasite survival with host pathology (406). This is exemplified through the theory of concomitant immunity where adult schistosomes protect the host from incoming larval stages of the same species for their own benefit (407). How this balance is affected when other pathogens are present is poorly understood.

Through the literature we found that Schistosoma spp., when co-infecting with other schistosomes, led to higher infection intensities, ectopic egg elimination (23, 53, 58, 59, 61, 64), and an increased risk of species hybridization which could enhance transmission (64, 7176). Outcomes of Schistosoma and STH co-infections are more complex, as both worsened and improved outcomes have been shown (7, 34). While parasite burden was unchanged in some cases, in others, liver pathology and egg granuloma sizes were both increased (7, 8689).

With most protozoan parasites, which require TH1 immune responses for protection, schistosomiasis-induced TH2 immune responses often led to worse prognoses. Cutaneous leishmaniasis required longer healing times post-treatment, and higher parasite burdens were observed in co-infected mice (134). Co-infection with trypanosomes generated increased levels of Trypanosoma parasitemia and Schistosoma egg-induced granulomas (144, 145). Toxoplasma gondii reduced helminth TH2 responses resulting in increased liver pathology and animal mortality (171). Finally, Entamoeba and Schistosoma co-infections yielded worsened parasite burdens of E. histolytica and animal morbidity (7, 190).

All the bacterial co-infections reviewed in this paper demonstrated that when present with schistosomiasis, outcomes were worse. Schistosomiasis is acknowledged to push latent Mycobacterium tuberculosis to reactivation (37) and may increase the transmission of Mycobacterium leprae (219221). While schistosomiasis parasite burden is lower during Salmonella co-infection, the risk of renal complications is increased (237, 242, 243) and antibiotic efficacy is reduced until anthelmintics are administered (235, 239, 246). Schistosomiasis, particularly S. haematobium, has also been seen to increase the risk of urinary tract infections by several bacterial species (259, 262, 264, 265, 271277).

Most viruses discussed here also led to worse outcomes when co-infecting with schistosomiasis. Schistosomiasis reduced protective TH1 immunity from HIV (325), resulting in increased HIV risk and transmission (36, 317), which is of great significance in cases of female genital schistosomiasis (323). Hepatitis viruses and schistosomiasis generally led to severe liver pathology and mortality (354, 361). Although research is limited, schistosomiasis may also impact the progression of CMV and arboviruses which are common in schistosomiasis endemic regions.

Intriguingly, when co-infecting with schistosomiasis, only one parasite and one viral pathogen created a positive outcome for the host, being malaria and SARS-CoV-2, respectively. In cases of Plasmodium and Schistosoma co-infection, despite an increased presentation of anemia and hematuria (100103, 105, 106), malaria parasite burden was reduced resulting in better outcomes. In the case of SARS-CoV-2, regression modeling of an observational cohort study implied that TH2-inducing parasites, including Schistosoma, decreased the risk of severe viral disease caused by immune hyperactivation, albeit this has yet to be confirmed in epidemiological data.

Despite the contentious data from Schistosoma and STH co-infections, and the amelioration of disease in Schistosoma co-infections with Plasmodium and possibly SARS-CoV-2, co-infections with schistosomiasis generally led to worsened outcomes as outlined in Table 1.

TABLE 1.

Schistosoma spp. and co-infection summary table

Co-infection with Schistosoma spp. Immunological outcomes Pathological outcomes Pathogen outcomes
Schistosoma Increased TH2 responses (56) Decreased liver pathology (23)
Increased bladder pathology (63)
Ectopic egg elimination (23, 53, 58, 59, 61)
Increased parasite burdens (23, 53, 55, 61, 68)
Soil-transmitted helminths Increased TH2 and TREG responses (84) and serum IgE (85) Pathology changes dependent on co-infecting STH Varying infection burdens depending on various factors
Plasmodium Increased anti-malaria IgG, IL-4 (109111), and TREG cells (114) Increased risk of anemia (100103, 105, 106) Decreased Plasmodium burden (102, 104, 115)
Leishmania Increased TH2 responses: IgE, IL-4, IL-5 (129)
Decreased TH1 responses in lymph nodes: IFNγ, NO, TNFα (mice) (134)
Smaller CL lesions (humans) (129) and larger lesions (mice) (134) which took longer to heal Increased levels of L. donovani amastigotes (mice) (135)
Trypanosoma Increased IL-10 (145)
Reduced IFNγ (145)
Splenomegaly (144)
Increased number of liver granulomas, with
higher cellularity and less fibrosis (145)
Higher mortality (144)
Increased Trypanosoma parasitemia (144, 145)
Decreased Schistosoma burden (7)
Toxoplasma Increased serum IgE and TNFα (169, 170)
Suppressed protective TH2 responses in livers (171)
Increased mortality when Toxoplasma
followed Schistosoma infection (169, 170)
Decreased Toxoplasma intestinal pathology (171)
Coagulative necrosis with extensive
hepatocyte vacuolization in livers (171)
Entamoeba Higher morbidity (7) Higher Entamoeba burdens (7)
Mycobacteria Increased TH2 responses, type 2 granulomas, IL-4204, TREG cells (206)
Dampened γδ T cell immune responses (218)
Reduction of TH1 CD4+ T cells (208)
Increased risk of clinical TB (196), TB reactivation (196, 197), and disseminated infection (196)
Increased risk of leprosy transmission (M. leprae) (219221)
Salmonella Increased anti-fibrotic TH1 responses in the liver, IL-10 (249), IFNγ (253)
Decreased type 2 mediators: IL-4 (249, 253), IL-5 (249)
Prolonged fever and hepatosplenomegaly (235)
Increased mortality (hamsters) (252), renal pathology (237, 242, 243), and liver pathology; egg granulomas were congested and necrotic (249)
Decreased effectiveness of antibiotic treatment (246)
Enhanced Salmonella colonization (235, 247249)
Lowered Schistosoma worm and egg burden (249, 253)
Urinary tract infection-causing agents Increased IL-4 (385)
Reduced invariant NK T cell-mediated killing (286) and antimicrobial peptides in the bladder (287)
Increased risk of UTI acquisition
(259, 262, 264, 265, 270277)
Severe acute respiratory syndrome coronavirus 2 Dampened immune hyperactivation (303) Potential reduced risk of severe disease (303)
Human immunodeficiency virus Increased TH2 responses (326, 327)
Decreased TH1 responses and protective CD8+ cells (325)
Increased HIV risk and transmission (36, 317)
Female genital schistosomiasis and HIV can lead to genital pathologies and infertility (323)
May lead to liver ailments (333): periportal fibrosis (334) and higher hepatotoxicity (335)
Potential for reduced egg excretion in feces (315, 336, 337)
HIV progression was not worse in co-infected (330, 331)
Hepatitis B virus and hepatitis C virus Increased TH2 responses: IL-4, IL-10 (354)
Decreased cell-mediated immune responses (364)
Clinical course of illness becomes more
severe (HBV and HCV) (354, 361): liver fibrosis and mortality (372, 373),
incidence of cirrhosis/hepatocellular carcinoma (367, 371)
Increased HCV RNA titers (367)

Ultimately, co-infections with schistosomiasis are underreported, understudied, and undertreated to an even greater extent than mono-infections with neglected tropical diseases. Yet these co-infections involve complex interactions of immunological factors and changes in pathology, often resulting in exacerbated disease. Research into Schistosoma co-infections, as demonstrated in this review, is beginning to describe these interactions. However, comprehensive epidemiological data are lacking, highlighting the need for larger cohort studies to determine the immunology of co-infecting pathogens. It is essential for the scientific research community to prioritize and expand this research further to better understand transmission kinetics, treatment, and vaccination challenges associated with co-infections. Acknowledging the reality of co-infections, and their immunological and pathological effects on each other, is crucial to improving patient care and developing effective intervention strategies in the coming years.

ACKNOWLEDGMENTS

We would like to thank Dr. Greg Matlashewski for his contributions editing this review. This work was supported by the Canadian Institutes of Health Research and the Fond de Recherche du Quebec en Santé.

The National Reference Centre for Parasitology is supported by the Public Health Agency of Canada/National Microbiology Laboratory, the Foundation of the Montreal General Hospital, the Foundation of the MUHC, the Research Institute of the MUHC, and the R. Howard Webster Foundation.

Biographies

graphic file with name cmr.00098-23.f007.gif

Dilhan Perera is completing his PhD in the division of Experimental Medicine, McGill University (Montreal, QC, Canada). Under the supervision of Dr. Momar Ndao, Dilhan’s current research focus is vaccine development and immunology for schistosomiasis and COVID-19. Dilhan earned his undergraduate degree in Microbiology and Immunology at McGill, and has spent time as a research assistant at the Mayo Clinic (Rochester, MN, USA) and KGK Science (London, ON, Canada) conducting in silico analyses of aneurysms and assisting with nutraceutical clinical trials, respectively. Since 2017, he has published eleven peer-reviewed manuscripts, both reviews and primary research, and serves as the secretary for the Food and Environmental Parasitology Network (Health Canada). Dilhan currently holds a Canadian Institutes of Health Research Doctoral Award, and has been awarded with several institutional presentation awards and awards dedicated to teaching, mentorship, and enhancing student learning.

graphic file with name cmr.00098-23.f008.gif

Cal Koger-Pease is completing their PhD in the division of Experimental Medicine, McGill University (Montreal, QC, Canada). Under the supervision of Dr. Momar Ndao, Cal’s current research focus is developing viral vectored vaccines and elucidating correlates of protection for schistosomiasis. Cal earned their undergraduate degree in Microbiology and Immunology at McGill, where they completed an undergraduate honours thesis focusing on short interfering RNA treatments for COVID-19. Since 2023, they have published two peer-reviewed manuscripts.

graphic file with name cmr.00098-23.f009.gif

Kayla Paulini is completing her PhD in the department of Microbiology and Immunology at McGill University (Montreal, QC, Canada). Under the supervision of Dr. Greg Matlashewski, Kayla studies various phosphatases in Leishmania and how they pertain to infection. She earned her bachelor’s degree in Microbiology and Immunology at McGill University in 2017 after gaining research experience in three different labs working with Caulobacter crescentus, Pseudomonas aeruginosa, and Heligmosomoides bakeri. After her bachelor’s degree, Kayla worked as a lab technician in the food microbiology industry before returning to graduate school. Currently, Kayla holds a Fond de Recherche du Québec en Santé doctoral award.

graphic file with name cmr.00098-23.f010.gif

Mohamed Daoudi is a post-doctoral fellow in the Ndao lab in the department of Microbiology and Immunology at McGill University (Montreal, QC, Canada). He holds a PhD in Microbiology and Medical Entomology focusing on investigating geographical distribution and vectorial capacity of the Moroccan population of sand flies. His current research project focuses on the utilisation of extracellular vesicles to develop advanced methods for the early detection of infections, the creation of precise diagnostic tools, and the preventive measures against Tick-borne diseases and schistosomiasis. Mohamed has published nine peer-reviewed manuscripts since 2019, received the Federation of African Immunological Societies Award in 2019, and received a grant from the Italian Ministry of Foreign Affairs and International Cooperation in 2020.

graphic file with name cmr.00098-23.f011.gif

Momar Ndao is a Doctor of Veterinary Medicine, and earned his MSc and PhD in Tropical Medicine in International Health and Tropical Diseases at the Institute of Tropical Medicine (Antwerp, Belgium). He is currently an Associate Professor at McGill University in the Faculty of Medicine (Montreal, QC, Canada) and his research focuses on parasite vaccinology, diagnostics, and biomarker discovery using proteomic analyses. Dr. Ndao serves as the Director of the Canadian National Reference Centre for Parasitology (NRCP) and holds the following appointments: Chair of the Facility Animal Care Committee (FACC) Research Institute of the McGill University Health Centre Glen, Co-chair of the Food and Environmental Parasitology Network (Health Canada), among others. He is also a member of several boards and committees including: the World Federation of Parasitologists, the executive board of the International Federation of Tropical Medicine, and the Scientific Committee of Pasteur Institute, to name a few.

Contributor Information

Momar Ndao, Email: momar.ndao@mcgill.ca.

Louisa A. Messenger, University of Nevada Las Vegas, Las Vegas, Nevada, USA

REFERENCES

  • 1. Schistosomiasis (Bilharzia). Available from: https://www.who.int/health-topics/schistosomiasis#tab=tab_1
  • 2. CDC - schistosomiasis. Available from: https://www.cdc.gov/parasites/schistosomiasis/index.html
  • 3. Colley DG, Bustinduy AL, Secor WE, King CH. 2014. Human schistosomiasis. Lancet 383:2253–2264. doi: 10.1016/S0140-6736(13)61949-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Pearce EJ, MacDonald AS. 2002. The immunobiology of schistosomiasis. Nat Rev Immunol 2:499–511. doi: 10.1038/nri843 [DOI] [PubMed] [Google Scholar]
  • 5. Martins Silva CL. 2016. Endothelial cells as targets of the intravascular parasitic disease schistosomiasis. Vascular Responses to Pathogens:195–207. doi: 10.1016/B978-0-12-801078-5.00015-7 [DOI] [Google Scholar]
  • 6. Abdel Aziz N, Musaigwa F, Mosala P, Berkiks I, Brombacher F. 2022. Type 2 immunity: a two-edged sword in schistosomiasis immunopathology. Trends Immunol 43:657–673. doi: 10.1016/j.it.2022.06.005 [DOI] [PubMed] [Google Scholar]
  • 7. Abruzzi A, Fried B. 2011. Coinfection of Schistosoma (Trematoda) with bacteria, protozoa and helminths. Adv Parasitol 77:1–85. doi: 10.1016/B978-0-12-391429-3.00005-8 [DOI] [PubMed] [Google Scholar]
  • 8. Schistosomiasis. https://www.who.int/en/news-room/fact-sheets/detail/schistosomiasis.
  • 9. Aula OP, McManus DP, Jones MK, Gordon CA. 2021. Schistosomiasis with a focus on Africa. Trop Med Infect Dis 6:109. doi: 10.3390/tropicalmed6030109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. van der Werf MJ, de Vlas SJ, Brooker S, Looman CWN, Nagelkerke NJD, Habbema JDF, Engels D. 2003. Quantification of clinical morbidity associated with schistosome infection in sub-Saharan Africa. Acta Trop 86:125–139. doi: 10.1016/s0001-706x(03)00029-9 [DOI] [PubMed] [Google Scholar]
  • 11. Thomas CM, Timson DJ. 2020. The mechanism of action of praziquantel: can new drugs exploit similar mechanisms? Curr Med Chem 27:676–696. doi: 10.2174/0929867325666180926145537 [DOI] [PubMed] [Google Scholar]
  • 12. Kura K, Ayabina D, Hollingsworth TD, Anderson RM. 2022. Determining the optimal strategies to achieve elimination of transmission for Schistosoma mansoni. Parasit Vectors 15:55. doi: 10.1186/s13071-022-05178-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Toor J, Alsallaq R, Truscott JE, Turner HC, Werkman M, Gurarie D, King CH, Anderson RM. 2018. Are we on our way to achieving the 2020 goals for schistosomiasis morbidity control using current world health organization guidelines Clin Infect Dis 66:S245–S252. doi: 10.1093/cid/ciy001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. King CH. 2015. It’s time to dispel the myth of “asymptomatic” schistosomiasis. PLoS Negl Trop Dis 9:e0003504. doi: 10.1371/journal.pntd.0003504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Eveland LK, Morse SI. 1978. Schistosoma mansoni: infectivity and immunizing effects of in vitro derived schistosomula attenuated by X irradiation. Exp Parasitol 45:19–25. doi: 10.1016/0014-4894(78)90040-1 [DOI] [PubMed] [Google Scholar]
  • 16. Minard P, Dean DA, Jacobson RH, Vannier WE, Murrell KD. 1978. Immunization of mice with cobalt-60 irradiated Schistosoma Mansoni cercariae. Am J Trop Med Hyg 27:76–86. doi: 10.4269/ajtmh.1978.27.76 [DOI] [PubMed] [Google Scholar]
  • 17. Perera D. J., Ndao M. 2021. Promising technologies in the field of helminth vaccines. Front Immunol 12:711650. doi: 10.3389/fimmu.2021.711650 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Perera DJ, Hassan AS, Jia Y, Ricciardi A, McCluskie MJ, Weeratna RD, Ndao M. 2020. Adjuvanted Schistosoma mansoni-cathepsin B With sulfated lactosyl archaeol archaeosomes or AddaVaxTM provides protection in a pre-clinical schistosomiasis Model. Front Immunol 11:605288. doi: 10.3389/fimmu.2020.605288 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Perera DJ, Hassan AS, Liu SS, Elahi SM, Gadoury C, Weeratna RD, Gilbert R, Ndao M. 2022. A low dose adenovirus vectored vaccine expressing Schistosoma mansoni cathepsin B protects from intestinal schistosomiasis in mice. EBioMedicine 80:104036. doi: 10.1016/j.ebiom.2022.104036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Molehin AJ. 2020. Schistosomiasis vaccine development: update on human clinical trials. J Biomed Sci 27:28. doi: 10.1186/s12929-020-0621-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Gryseels B, Polman K, Clerinx J, Kestens L. 2006. Human schistosomiasis. Lancet 368:1106–1118. doi: 10.1016/S0140-6736(06)69440-3 [DOI] [PubMed] [Google Scholar]
  • 22. De Clercq D, Vercruysse J, Picquet M, Shaw DJ, Diop M, Ly A, Gryseels B. 1999. The epidemiology of a recent focus of mixed Schistosoma haematobium and Schistosoma mansoni infections around the ‘Lac de Guiers’ in the Senegal River Basin, Senegal. Trop Med Int Health 4:544–550. doi: 10.1046/j.1365-3156.1999.00444.x [DOI] [PubMed] [Google Scholar]
  • 23. Cunin P, Tchuem Tchuenté LA, Poste B, Djibrilla K, Martin PMV. 2003. Interactions between Schistosoma haematobium and Schistosoma mansoni in humans in north Cameroon. Trop Med Int Health 8:1110–1117. doi: 10.1046/j.1360-2276.2003.01139.x [DOI] [PubMed] [Google Scholar]
  • 24. De Leo GA, Stensgaard A-S, Sokolow SH, N’Goran EK, Chamberlin AJ, Yang G-J, Utzinger J. 2020. Schistosomiasis and climate change. BMJ 371:m4324. doi: 10.1136/bmj.m4324 [DOI] [Google Scholar]
  • 25. Picquet M, Ernould JC, Vercruysse J, Southgate VR, Mbaye A, Sambou B, Niang M, Rollinson D. 1996. The epidemiology of human schistosomiasis in the Senegal river Basin. Transactions of the Royal Society of Tropical Medicine and Hygiene 90:340–346. doi: 10.1016/S0035-9203(96)90501-5 [DOI] [PubMed] [Google Scholar]
  • 26. Chaine JP, Malek EA. 1983. Urinary schistosomiasis in the sahelian region of the Senegal river basin. Trop Geogr Med 35:249–256. [PubMed] [Google Scholar]
  • 27. Talla I, Kongs A, Verlé P, Belot J, Sarr S, Coll AM. 1990. Outbreak of intestinal schistosomiasis in the Senegal River Basin. Ann Soc Belg Med Trop 70:173–180. [PubMed] [Google Scholar]
  • 28. Talla I, Kongs A, Verlé P. 1992. Preliminary study of the prevalence of human schistosomiasis in Richard-Toll (the Senegal river basin). Trans R Soc Trop Med Hyg 86:182. doi: 10.1016/0035-9203(92)90562-q [DOI] [PubMed] [Google Scholar]
  • 29. Boissier J, Grech-Angelini S, Webster BL, Allienne J-F, Huyse T, Mas-Coma S, Toulza E, Barré-Cardi H, Rollinson D, Kincaid-Smith J, Oleaga A, Galinier R, Foata J, Rognon A, Berry A, Mouahid G, Henneron R, Moné H, Noel H, Mitta G. 2016. Outbreak of urogenital schistosomiasis in Corsica (France): an epidemiological case study. Lancet Infect Dis 16:971–979. doi: 10.1016/S1473-3099(16)00175-4 [DOI] [PubMed] [Google Scholar]
  • 30. Rothe C, Zimmer T, Schunk M, Wallrauch C, Helfrich K, Gültekin F, Bretzel G, Allienne J-F, Boissier J. 2021. Developing endemicity of schistosomiasis, Corsica, France. Emerg Infect Dis 27:319–321. doi: 10.3201/eid2701.204391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Wellinghausen N, Moné H, Mouahid G, Nebel A, Tappe D, Gabriel M. 2022. A family cluster of schistosomiasis acquired in Solenzara River, Corsica (France) — Solenzara River is clearly a transmission site for schistosomiasis in Corsica. Parasitol Res 121:2449–2452. doi: 10.1007/s00436-022-07574-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Léger E, Borlase A, Fall CB, Diouf ND, Diop SD, Yasenev L, Catalano S, Thiam CT, Ndiaye A, Emery A, Morrell A, Rabone M, Ndao M, Faye B, Rollinson D, Rudge JW, Sène M, Webster JP. 2020. Prevalence and distribution of schistosomiasis in human, livestock, and snail populations in northern Senegal: a One Health epidemiological study of a multi-host system. Lancet Planet Health 4:e330–e342. doi: 10.1016/S2542-5196(20)30129-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Stensgaard AS, Vounatsou P, Sengupta ME, Utzinger J. 2019. Schistosomes, snails and climate change: current trends and future expectations. Acta Trop 190:257–268. doi: 10.1016/j.actatropica.2018.09.013 [DOI] [PubMed] [Google Scholar]
  • 34. Geiger SM. 2008. Immuno-epidemiology of Schistosoma mansoni infections in endemic populations co-infected with soil-transmitted helminths: present knowledge, challenges, and the need for further studies. Acta Trop 108:118–123. doi: 10.1016/j.actatropica.2008.05.012 [DOI] [PubMed] [Google Scholar]
  • 35. Phillips MA, Burrows JN, Manyando C, van Huijsduijnen RH, Van Voorhis WC, Wells TNC. 2017. Malaria. Nat Rev Dis Primers 3:17050. doi: 10.1038/nrdp.2017.50 [DOI] [PubMed] [Google Scholar]
  • 36. Patel P, Rose CE, Kjetland EF, Downs JA, Mbabazi PS, Sabin K, Chege W, Watts DH, Secor WE. 2021. Association of schistosomiasis and HIV infections: a systematic review and meta-analysis. Int J Infect Dis 102:544–553. doi: 10.1016/j.ijid.2020.10.088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Baya B, Kone B, Somboro A, Kodio O, Somboro AM, Diarra B, Traore FG, Kone D, Traore MA, Kone M, Togo AG, Sarro YS, Maiga A, Maiga M, Toloba Y, Diallo S, Murphy RL, Doumbia S. 2023. Prevalence and clinical relevance of Schistosoma mansoni co-infection with Mycobacterium tuberculosis: a systematic literature review. Open J Epidemiol 13:97–111. doi: 10.4236/ojepi.2023.131008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Petrellis G, Piedfort O, Katsandegwaza B, Dewals BG. 2023. Parasitic worms affect virus coinfection: a mechanistic overview. Trends Parasitol 39:358–372. doi: 10.1016/j.pt.2023.02.007 [DOI] [PubMed] [Google Scholar]
  • 39. Rovira-Diaz E, El-Naccache DW, Reyes J, Zhao Y, Nasuhidehnavi A, Chen F, Gause WC, Yap GS. 2022. The impact of helminth coinfection on innate and adaptive immune resistance and disease tolerance during toxoplasmosis. J Immunol 209:2160–2171. doi: 10.4049/jimmunol.2200504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Desai P, Diamond MS, Thackray LB. 2021. Helminth-virus interactions: determinants of coinfection outcomes. Gut Microbes 13:1961202. doi: 10.1080/19490976.2021.1961202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Winkel BMF, Dalenberg MR, de Korne CM, Feijt C, Langenberg MCC, Pelgrom L, Ganesh MS, Yazdanbakhsh M, Smits HH, de Jong EC, Everts B, van Leeuwen FWB, Hokke CH, Roestenberg M. 2018. Early induction of human regulatory dermal antigen presenting cells by skin-penetrating Schistosoma mansoni cercariae. Front Immunol 9:2510. doi: 10.3389/fimmu.2018.02510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. de Jesus AR, Silva A, Santana LB, Magalhães A, de Jesus AA, de Almeida RP, Rêgo MAV, Burattini MN, Pearce EJ, Carvalho EM. 2002. Clinical and immunologic evaluation of 31 patients with acute schistosomiasis mansoni. J Infect Dis 185:98–105. doi: 10.1086/324668 [DOI] [PubMed] [Google Scholar]
  • 43. Ross AG, Vickers D, Olds GR, Shah SM, McManus DP. 2007. Katayama syndrome. Lancet Infect Dis 7:218–224. doi: 10.1016/S1473-3099(07)70053-1 [DOI] [PubMed] [Google Scholar]
  • 44. Schwartz E, Rozenman J, Perelman M. 2000. Pulmonary manifestations of early schistosome infection among nonimmune travelers. Am J Med 109:718–722. doi: 10.1016/s0002-9343(00)00619-7 [DOI] [PubMed] [Google Scholar]
  • 45. Pearce EJ, M Kane C, Sun J, J Taylor J, McKee AS, Cervi L. 2004. Th2 response polarization during infection with die helminth parasite Schistosoma mansoni. Immunol Rev 201:117–126. doi: 10.1111/j.0105-2896.2004.00187.x [DOI] [PubMed] [Google Scholar]
  • 46. Brunet LR, Finkelman FD, Cheever AW, Kopf MA, Pearce EJ. 1997. IL-4 protects against TNF-alpha-mediated cachexia and death during acute schistosomiasis. J Immunol 159:777–785. doi: 10.4049/jimmunol.159.2.777 [DOI] [PubMed] [Google Scholar]
  • 47. Schwartz C, Fallon PG. 2018. Schistosoma “eggs-iting” the host: granuloma formation and egg excretion. Front Immunol 9:2492. doi: 10.3389/fimmu.2018.02492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Tawiah D. 2014. Female genital schistosomiasis, genital tract infections and HIV co-infection in the Volta Basin of Ghana. IJTDH 3:94–103. doi: 10.9734/IJTDH/2013/1010 [DOI] [Google Scholar]
  • 49. Feldmeier H, Krantz I, Poggensee G. 1994. Female genital schistosomiasis as a risk-factor for the transmission of HIV. Int J STD AIDS 5:368–372. doi: 10.1177/095646249400500517 [DOI] [PubMed] [Google Scholar]
  • 50. Sturt AS, Webb EL, Francis SC, Hayes RJ, Bustinduy AL. 2020. Beyond the barrier: female genital schistosomiasis as a potential risk factor for HIV-1 acquisition. Acta Trop 209:105524. doi: 10.1016/j.actatropica.2020.105524 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Kjetland EF, Ndhlovu PD, Mduluza T, Deschoolmeester V, Midzi N, Gomo E, Gwanzura L, Mason PR, Vermorken JB, Friis H, Gundersen SG, Baay MFD. 2010. The effects of genital Schistosoma haematobium on human papillomavirus and the development of cervical neoplasia after five years in a Zimbabwean population. Eur J Gynaecol Oncol 31:169–173. [PubMed] [Google Scholar]
  • 52. Mohammed SA, Hetta HF, Zahran AM, Tolba MEM, Attia RAH, Behnsawy HM, Algammal AM, Batiha G-S, Mohammed AQ, Ahmad AA. 2023. T cell subsets, regulatory T, regulatory B cells and proinflammatory cytokine profile in Schistosoma haematobium associated bladder cancer: first report from Upper Egypt. PLoS Negl Trop Dis 17:e0011258. doi: 10.1371/journal.pntd.0011258 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Meurs L, Mbow M, Vereecken K, Menten J, Mboup S, Polman K. 2012. Epidemiology of mixed Schistosoma mansoni and Schistosoma haematobium infections in northern Senegal. Int J Parasitol 42:305–311. doi: 10.1016/j.ijpara.2012.02.002 [DOI] [PubMed] [Google Scholar]
  • 54. Webster BL, Diaw OT, Seye MM, Faye DS, Stothard JR, Sousa-Figueiredo JC, Rollinson D. 2013. Praziquantel treatment of school children from single and mixed infection foci of intestinal and urogenital schistosomiasis along the Senegal River Basin: monitoring treatment success and re-infection patterns. Acta Trop 128:292–302. doi: 10.1016/j.actatropica.2012.09.010 [DOI] [PubMed] [Google Scholar]
  • 55. Knowles SCL, Webster BL, Garba A, Sacko M, Diaw OT, Fenwick A, Rollinson D, Webster JP. 2015. Epidemiological interactions between urogenital and intestinal human schistosomiasis in the context of praziquantel treatment across three West African countries. PLoS Negl Trop Dis 9:e0004019. doi: 10.1371/journal.pntd.0004019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Meurs L, Mbow M, Boon N, Vereecken K, Amoah AS, Labuda LA, Dièye TN, Mboup S, Yazdanbakhsh M, Polman K. 2014. Cytokine responses to Schistosoma mansoni and Schistosoma haematobium in relation to infection in a co-endemic focus in northern Senegal. PLoS Negl Trop Dis 8:e3080. doi: 10.1371/journal.pntd.0003080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Meurs L, Mbow M, Boon N, van den Broeck F, Vereecken K, Dièye TN, Abatih E, Huyse T, Mboup S, Polman K. 2013. Micro-geographical heterogeneity in Schistosoma mansoni and S. haematobium infection and morbidity in a co-endemic community in northern Senegal. PLoS Negl Trop Dis 7:e2608. doi: 10.1371/journal.pntd.0002608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Meurs L, Mbow M, Vereecken K, Menten J, Mboup S, Polman K. 2012. Bladder morbidity and hepatic fibrosis in mixed Schistosoma haematobium and S. mansoni infections: a population-wide study in Northern Senegal. PLoS Negl Trop Dis 6:e1829. doi: 10.1371/journal.pntd.0001829 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Cunin P, Griffet A, Poste B, Djibrilla K, Martin PMV. 2000. Epidemic Schistosoma mansoni in a known S. haematobium area. Trans R Soc Trop Med Hyg 94:657–660. doi: 10.1016/s0035-9203(00)90221-9 [DOI] [PubMed] [Google Scholar]
  • 60. Ugbomoiko US, Dalumo V, Danladi YK, Heukelbach J, Ofoezie IE. 2012. Concurrent urinary and intestinal schistosomiasis and intestinal helminthic infections in schoolchildren in Ilobu, South-western Nigeria. Acta Trop 123:16–21. doi: 10.1016/j.actatropica.2012.03.002 [DOI] [PubMed] [Google Scholar]
  • 61. Anyan WK, Abonie SD, Aboagye-Antwi F, Tettey MD, Nartey LK, Hanington PC, Anang AK, Muench SB. 2019. Concurrent Schistosoma mansoni and Schistosoma haematobium infections in a peri-urban community along the Weija dam in Ghana: a wake up call for effective National Control Programme. Acta Trop 199:105116. doi: 10.1016/j.actatropica.2019.105116 [DOI] [PubMed] [Google Scholar]
  • 62. Gouvras AN, Kariuki C, Koukounari A, Norton AJ, Lange CN, Ireri E, Fenwick A, Mkoji GM, Webster JP. 2013. The impact of single versus mixed Schistosoma haematobium and S. mansoni infections on morbidity profiles amongst school-children in Taveta, Kenya. Acta Trop 128:309–317. doi: 10.1016/j.actatropica.2013.01.001 [DOI] [PubMed] [Google Scholar]
  • 63. Koukounari A, Donnelly CA, Sacko M, Keita AD, Landouré A, Dembelé R, Bosqué-Oliva E, Gabrielli AF, Gouvras A, Traoré M, Fenwick A, Webster JP. 2010. The impact of single versus mixed schistosome species infections on liver, spleen and bladder morbidity within Malian children pre- and post-praziquantel treatment. BMC Infect Dis 10:227. doi: 10.1186/1471-2334-10-227 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Fall CB, Lambert S, Léger E, Yasenev L, Garba AD, Diop SD, Borlase A, Catalano S, Faye B, Walker M, Sene M, Webster JP. 2021. Hybridized zoonotic Schistosoma infections result in hybridized morbidity profiles: a clinical morbidity study amongst co-infected human populations of Senegal. Microorganisms 9:1776. doi: 10.3390/microorganisms9081776 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Webster BL, Southgate VR, Tchuem Tchuenté LA. 1999. Mating interactions between Schistosoma haematobium and S. mansoni. J Helminthol 73:351–356. [PubMed] [Google Scholar]
  • 66. Brumpt E. 1930. Localisation vésicale expérimentale d’œufs de Schistosoma mansoni chez une souris. Localisations vésicales à S. mansoni et rectales à S. haematobium chez l’homme. Ann Parasitol Hum Comp 8:298–308. doi: 10.1051/parasite/1930083298 [DOI] [Google Scholar]
  • 67. Barsoum RS, Esmat G, El-Baz T. 2013. Human schistosomiasis: clinical perspective: review. J Adv Res 4:433–444. doi: 10.1016/j.jare.2013.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Garba A, Lamine MS, Barkiré N, Djibo A, Sofo B, Gouvras AN, Labbo R, Sebangou H, Webster JP, Fenwick A, Utzinger J. 2013. Efficacy and safety of two closely spaced doses of praziquantel against Schistosoma haematobium and S. mansoni and re-infection patterns in school-aged children in Niger. Acta Trop 128:334–344. doi: 10.1016/j.actatropica.2012.08.008 [DOI] [PubMed] [Google Scholar]
  • 69. Tchuem Tchuenté L-A, Momo SC, Stothard JR, Rollinson D. 2013. Efficacy of praziquantel and reinfection patterns in single and mixed infection foci for intestinal and urogenital schistosomiasis in Cameroon. Acta Trop 128:275–283. doi: 10.1016/j.actatropica.2013.06.007 [DOI] [PubMed] [Google Scholar]
  • 70. Odegaard JI, Hsieh MH. 2014. Immune responses to Schistosoma haematobium infection. Parasite Immunol 36:428–438. doi: 10.1111/pim.12084 [DOI] [PubMed] [Google Scholar]
  • 71. Panzner U, Boissier J. 2021. Natural intra- and interclade human hybrid schistosomes in Africa with considerations on prevention through vaccination. Microorganisms 9:1465. doi: 10.3390/microorganisms9071465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Leger E, Webster JP. 2017. Hybridizations within the Genus Schistosoma: implications for evolution, epidemiology and control. Parasitology 144:65–80. doi: 10.1017/S0031182016001190 [DOI] [PubMed] [Google Scholar]
  • 73. King KC, Stelkens RB, Webster JP, Smith DF, Brockhurst MA. 2015. Hybridization in parasites: consequences for adaptive evolution, pathogenesis, and public health in a changing world. PLoS Pathog 11:e1005098. doi: 10.1371/journal.ppat.1005098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Depaquit J, Akhoundi M, Haouchine D, Mantelet S, Izri A. 2019. No limit in interspecific hybridization in schistosomes: observation from a case report. Parasite 26:10. doi: 10.1051/parasite/2019010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Le Govic Y, Kincaid-Smith J, Allienne J-F, Rey O, de Gentile L, Boissier J. 2019. Schistosoma haematobium–Schistosoma mansoni hybrid parasite in migrant boy. Emerg Infect Dis 25:365–367. doi: 10.3201/eid2502.172028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Huyse T, Van den Broeck F, Hellemans B, Volckaert FAM, Polman K. 2013. Hybridisation between the two major African schistosome species of humans. Int J Parasitol 43:687–689. doi: 10.1016/j.ijpara.2013.04.001 [DOI] [PubMed] [Google Scholar]
  • 77. Soil-transmitted helminth infections. https://www.who.int/news-room/fact-sheets/detail/soil-transmitted-helminth-infections.
  • 78. Jourdan PM, Lamberton PHL, Fenwick A, Addiss DG. 2018. Soil-transmitted helminth infections. Lancet 391:252–265. doi: 10.1016/S0140-6736(17)31930-X [DOI] [PubMed] [Google Scholar]
  • 79. Vacca F, Le Gros G. 2022. Tissue-specific immunity in helminth infections. Mucosal Immunol 15:1212–1223. doi: 10.1038/s41385-022-00531-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Maizels RM, Hewitson JP, Smith KA. 2012. Susceptibility and immunity to helminth parasites. Curr Opin Immunol 24:459–466. doi: 10.1016/j.coi.2012.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. McSorley HJ, Maizels RM. 2012. Helminth infections and host immune regulation. Clin Microbiol Rev 25:585–608. doi: 10.1128/CMR.05040-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Yajima A, Gabrielli AF, Montresor A, Engels D. 2011. Moderate and high endemicity of schistosomiasis is a predictor of the endemicity of soil-transmitted helminthiasis - systematic review. Trans R Soc Trop Med Hyg 105:68–73. doi: 10.1016/j.trstmh.2010.11.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Schistosomiasis and soil transmitted helminthiases: progress report, 2021 – Schistosomiase et Géohelminthiases: rapport de situation. 2021. Weekly Epidemiological Record = Relevé Épidémiologique Hebdomadaire 97, 621–631.
  • 84. Babu S, Nutman TB. 2019. 31 - immune responses to helminth infection, p 437–447. In Clinical immunology: principles and practice [Google Scholar]
  • 85. de Rezende MC, Araújo ES, Moreira JMP, Rodrigues VF, Rodrigues JL, Pereira C de J, Negrão-Corrêa D. 2015. Effect of different stages of Schistosoma mansoni infection on the parasite burden and immune response to Strongyloides venezuelensis in co-infected mice. Parasitol Res 114:4601–4616. doi: 10.1007/s00436-015-4706-6 [DOI] [PubMed] [Google Scholar]
  • 86. de Rezende MC, Moreira JMP, Fernandes LLM, Rodrigues VF, Negrão-Corrêa D. 2020. Strongyloides venezuelensis-infection alters the profile of cytokines and liver inflammation in mice co-infected with Schistosoma mansoni. Cytokine 127:154931. doi: 10.1016/j.cyto.2019.154931 [DOI] [PubMed] [Google Scholar]
  • 87. Bazzone LE, Smith PM, Rutitzky LI, Shainheit MG, Urban JF, Setiawan T, Blum AM, Weinstock JV, Stadecker MJ. 2008. Coinfection with the intestinal nematode Heligmosomoides polygyrus markedly reduces hepatic egg-induced immunopathology and proinflammatory cytokines in mouse models of severe schistosomiasis. Infect Immun 76:5164–5172. doi: 10.1128/IAI.00673-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Bickle QD, Solum J, Helmby H. 2008. Chronic intestinal nematode infection exacerbates experimental Schistosoma mansoni infection. Infect Immun 76:5802–5809. doi: 10.1128/IAI.00827-08 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Le L, Khatoon S, Jiménez P, Peterson C, Kernen R, Zhang W, Molehin AJ, Lazarus S, Sudduth J, May J, Karmakar S, Rojo JU, Ahmad G, Torben W, Carey D, Wolf RF, Papin JF, Siddiqui AA. 2020. Chronic whipworm infection exacerbates Schistosoma mansoni egg-induced hepatopathology in non-human primates. Parasit Vectors 13:109. doi: 10.1186/s13071-020-3980-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Geiger SM, Alexander NDE, Fujiwara RT, Brooker S, Cundill B, Diemert DJ, Correa-Oliveira R, Bethony JM. 2011. Necator americanus and helminth co-infections: further down-modulation of hookworm-specific type 1 immune responses. PLoS Negl Trop Dis 5:e1280. doi: 10.1371/journal.pntd.0001280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Adewale B, Heintz JR, Pastore CF, Rossi HL, Hung L-Y, Rahman N, Bethony J, Diemert D, Babatunde JA, Herbert DR. 2021. Parasitic helminth infections in humans modulate Trefoil Factor levels in a manner dependent on the species of parasite and age of the host. PLoS Negl Trop Dis 15:e0009550. doi: 10.1371/journal.pntd.0009550 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Pinot de Moira A, Jones FM, Wilson S, Tukahebwa E, Fitzsimmons CM, Mwatha JK, Bethony JM, Kabatereine NB, Dunne DW, Urban JF. 2013. Effects of treatment on IgE responses against parasite allergen-like proteins and immunity to reinfection in childhood schistosome and hookworm coinfections. Infect Immun 81:23–32. doi: 10.1128/IAI.00748-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Pinot de Moira A, Fitzsimmons CM, Jones FM, Wilson S, Cahen P, Tukahebwa E, Mpairwe H, Mwatha JK, Bethony JM, Skov PS, Kabatereine NB, Dunne DW. 2014. Suppression of basophil histamine release and other IgE-dependent responses in childhood Schistosoma mansoni/hookworm coinfection. J Infect Dis 210:1198–1206. doi: 10.1093/infdis/jiu234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Campolina SS, Araujo MSS, Rezende T, Matoso L, Quites HFO, Teixeira-Carvalho A, Martins-Filho OA, Gazzinelli A, Correa-Oliveira R. 2015. Effective anthelmintic therapy of residents living in endemic area of high prevalence for Hookworm and Schistosoma mansoni infections enhances the levels of allergy risk factor anti-Der p1 IgE. Results Immunol 5:6–12. doi: 10.1016/j.rinim.2013.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Malaria. https://www.who.int/news-room/fact-sheets/detail/malaria.
  • 96. Parums DV. 2023. Editorial: current status of two adjuvanted malaria vaccines and the world health organization (WHO) strategy to eradicate malaria by 2030. Med Sci Monit 29:e939357. doi: 10.12659/MSM.939357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. López C, Yepes-Pérez Y, Hincapié-Escobar N, Díaz-Arévalo D, Patarroyo MA. 2017. What is known about the immune response induced by Plasmodium vivax malaria vaccine candidates?. Front Immunol 8:126. doi: 10.3389/fimmu.2017.00126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. CDC - Schistosomiasis - Epidemiology & Risk Factors. https://www.cdc.gov/parasites/schistosomiasis/epi.html.
  • 99. Degarege A, Degarege D, Veledar E, Erko B, Nacher M, Beck-Sague CM, Madhivanan P. 2016. Plasmodium falciparum infection status among children with Schistosoma in sub-Saharan Africa: a systematic review and meta-analysis. PLoS Negl Trop Dis 10:e0005193. doi: 10.1371/journal.pntd.0005193 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Hailu T, Yimer M, Mulu W, Abera B. 2018. Synergetic effects of Plasmodium, hookworm, and Schistosoma mansoni infections on hemoglobin level among febrile school age children in Jawe Worda, Northwest Ethiopia. J Parasitol Res 2018:9573413. doi: 10.1155/2018/9573413 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Orish VN, Ofori-Amoah J, Amegan-Aho KH, Osei-Yeboah J, Lokpo SY, Osisiogu EU, Agordoh PD, Adzaku FK. 2019. Prevalence of polyparasitic infection among primary school children in the volta region of Ghana. Open Forum Infect Dis 6:ofz153. doi: 10.1093/ofid/ofz153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Sumbele IUN, Otia OV, Bopda OSM, Ebai CB, Kimbi HK, Nkuo-Akenji T. 2021. Polyparasitism with Schistosoma haematobium, Plasmodium and soil-transmitted helminths in school-aged children in Muyuka-Cameroon following implementation of control measures: a cross sectional study. Infect Dis Poverty 10:14. doi: 10.1186/s40249-021-00802-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Anchang-Kimbi JK, Elad DM, Sotoing GT, Achidi EA. 2017. Coinfection with Schistosoma haematobium and Plasmodium falciparum and anaemia severity among pregnant women in Munyenge, mount cameroon area: a cross-sectional study. J Parasitol Res 2017:6173465. doi: 10.1155/2017/6173465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. McDowell D, Hurt L, Kabatereine NB, Stothard JR, Lello J. 2022. Infection history and current coinfection with Schistosoma mansoni decreases Plasmodium species intensities in preschool children in Uganda. J Infect Dis 225:2181–2186. doi: 10.1093/infdis/jiac072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Tuasha N, Hailemeskel E, Erko B, Petros B. 2019. Comorbidity of intestinal helminthiases among malaria outpatients of Wondo Genet health centers, southern Ethiopia: implications for integrated control. BMC Infect Dis 19:659. doi: 10.1186/s12879-019-4290-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Nyarko R, Torpey K, Ankomah A. 2018. Schistosoma haematobium, Plasmodium falciparum infection and anaemia in children in Accra, Ghana. Trop Dis Travel Med Vaccines 4:3. doi: 10.1186/s40794-018-0063-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Dassah S, Asiamah GK, Harun V, Appiah-Kubi K, Oduro A, Asoala V, Amenga-Etego L. 2022. Urogenital schistosomiasis transmission, malaria and anemia among school-age children in Northern Ghana. Heliyon 8:e10440. doi: 10.1016/j.heliyon.2022.e10440 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Morenikeji OA, Eleng IE, Atanda OS, Oyeyemi OT. 2016. Renal related disorders in concomitant Schistosoma haematobium-Plasmodium falciparum infection among children in a rural community of Nigeria. J Infect Public Health 9:136–142. doi: 10.1016/j.jiph.2015.06.013 [DOI] [PubMed] [Google Scholar]
  • 109. Santano R, Rubio R, Grau-Pujol B, Escola V, Muchisse O, Cuamba I, Vidal M, Cisteró P, Ruiz-Olalla G, Aguilar R, et al. 2021. Plasmodium falciparum and helminth coinfections increase IGE and parasite-specific IgG responses. Microbiol Spectr 9:e0110921. doi: 10.1128/Spectrum.01109-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Tokplonou L, Nouatin O, Sonon P, M’po G, Glitho S, Agniwo P, Gonzalez-Ortiz D, Tchégninougbo T, Ayitchédji A, Favier B, Donadi EA, Milet J, Luty AJF, Massougbodji A, Garcia A, Ibikounlé M, Courtin D. 2020. Schistosoma haematobium infection modulates Plasmodium falciparum parasite density and antimalarial antibody responses. Parasite Immunol 42:e12702. doi: 10.1111/pim.12702 [DOI] [PubMed] [Google Scholar]
  • 111. Lyke KE, Dabo A, Arama C, Diarra I, Plowe CV, Doumbo OK, Sztein MB. 2017. Long-term maintenance of CD4 T cell memory responses to malaria antigens in Malian children coinfected with Schistosoma haematobium. Front Immunol 8:1995. doi: 10.3389/fimmu.2017.01995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Nyakundi RK, Nyamongo O, Maamun J, Akinyi M, Mulei I, Farah IO, Blankenship D, Grimberg B, Hau J, Malhotra I, Ozwara H, King CL, Kariuki TM. 2016. Protective effect of chronic schistosomiasis in baboons coinfected with Schistosoma mansoni and Plasmodium knowlesi. Infect Immun 84:1320–1330. doi: 10.1128/IAI.00490-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Nyakundi RK, Hau J, Ogongo P, Nyamongo O, Jeneby M, Akinyi M, Mulei I, Nyundo F, Farah I, Malhotra I, Ozwara H, King CL, Kariuki T. 2022. Acquired clinical immunity to malaria in nonhuman primates coinfected with Schistosoma and Plasmodium parasites. Infect Immun 90:e0046421. doi: 10.1128/IAI.00464-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Wang M, Feng Y, Pang W, Qi Z, Zhang Y, Guo Y, Luo E, Cao Y. 2014. Parasite densities modulate susceptibility of mice to cerebral malaria during co-infection with Schistosoma japonicum and Plasmodium berghei. Malar J 13:116. doi: 10.1186/1475-2875-13-116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Moriyasu T, Nakamura R, Deloer S, Senba M, Kubo M, Inoue M, Culleton R, Hamano S. 2018. Schistosoma mansoni infection suppresses the growth of Plasmodium yoelii parasites in the liver and reduces gametocyte infectivity to mosquitoes. PLoS Negl Trop Dis 12:e0006197. doi: 10.1371/journal.pntd.0006197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Abay SM, Tilahun M, Fikrie N, Habtewold A. 2013. Plasmodium falciparum and Schistosoma mansoni coinfection and the side benefit of artemether-lumefantrine in malaria patients. J Infect Dev Ctries 7:468–474. doi: 10.3855/jidc.2658 [DOI] [PubMed] [Google Scholar]
  • 117. Mbanzulu KM, Zanga JK, Mukendi JPK, Mbaya Ntita F, Matangila JR, Muhindo HM, Mpoyi Wa Mpoyi S, Aloni MN, Wumba R. 2018. The impact of artesunate-amodiaquine on Schistosoma mansoni infection among children infected by Plasmodium in rural area of lemfu, kongo central, democratic Republic of the Congo. Biomed Res Int 2018:3487183. doi: 10.1155/2018/3487183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Pishchany G, Skaar EP. 2012. Taste for blood: hemoglobin as a nutrient source for pathogens. PLoS Pathog 8:e1002535. doi: 10.1371/journal.ppat.1002535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Figueiredo B-P, Ricci ND, de Assis NRG, de Morais SB, Fonseca CT, Oliveira SC. 2015. Kicking in the guts: Schistosoma mansoni digestive tract proteins are potential candidates for vaccine development. Front Immunol 6:22. doi: 10.3389/fimmu.2015.00022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Coronado LM, Montealegre S, Chaverra Z, Mojica L, Espinosa C, Almanza A, Correa R, Stoute JA, Gittens RA, Spadafora C. 2016. Blood stage Plasmodium falciparum exhibits biological responses to direct current electric fields. PLoS One 11:e0161207. doi: 10.1371/journal.pone.0161207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Huang H-H, Rigouin C, Williams DL. 2012. The redox biology of schistosome parasites and applications for drug development. Curr Pharm Des 18:3595–3611. [PMC free article] [PubMed] [Google Scholar]
  • 122. Burza S, Croft SL, Boelaert M. 2018. Leishmaniasis. Lancet 392:951–970. doi: 10.1016/S0140-6736(18)31204-2 [DOI] [PubMed] [Google Scholar]
  • 123. Lypaczewski P, Matlashewski G. 2021. Leishmania donovani hybridisation and introgression in nature: a comparative genomic investigation. Lancet Microbe 2:e250–e258. doi: 10.1016/S2666-5247(21)00028-8 [DOI] [PubMed] [Google Scholar]
  • 124. Volpedo G, Huston RH, Holcomb EA, Pacheco-Fernandez T, Gannavaram S, Bhattacharya P, Nakhasi HL, Satoskar AR. 2021. From infection to vaccination: reviewing the global burden, history of vaccine development, and recurring challenges in global leishmaniasis protection. Expert Rev Vaccines 20:1431–1446. doi: 10.1080/14760584.2021.1969231 [DOI] [PubMed] [Google Scholar]
  • 125. Paulini K, Lypaczewski P, Zhang W-W, Perera DJ, Ndao M, Matlashewski G. 2022. Investigating the Leishmania donovani sacp gene and its role in macrophage infection and survival in mice. Trop Med Infect Dis 7:384. doi: 10.3390/tropicalmed7110384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Prevention C.-C. for D. C. and. CDC - Leishmaniasis - Biology. (2020).
  • 127. Miranda GS, Resende SD, Cardoso DT, Camelo GMA, Silva J, de Castro VN, Geiger SM, Carneiro M, Negrão-Corrêa D. 2021. Previous history of American tegumentary leishmaniasis alters susceptibility and immune response against Schistosoma mansoni infection in humans. Front Immunol 12:630934. doi: 10.3389/fimmu.2021.630934 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Carstens-Kass J, Paulini K, Lypaczewski P, Matlashewski G. 2021. A review of the leishmanin skin test: a neglected test for a neglected disease. PLoS Negl Trop Dis 15:e0009531. doi: 10.1371/journal.pntd.0009531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. O’Neal SE, Guimarães LH, Machado PR, Alcântara L, Morgan DJ, Passos S, Glesby MJ, Carvalho EM. 2007. Influence of helminth infections on the clinical course of and immune response to Leishmania braziliensis cutaneous leishmaniasis. J Infect Dis 195:142–148. doi: 10.1086/509808 [DOI] [PubMed] [Google Scholar]
  • 130. Fitzsimmons CM, Falcone FH, Dunne DW. 2014. Helminth allergens, parasite-specific IgE, and its protective role in human immunity. Front Immunol 5:61. doi: 10.3389/fimmu.2014.00061 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Geiger SM, Jardim-Botelho A, Williams W, Alexander N, Diemert DJ, Bethony JM. 2013. Serum CCL11 (eotaxin-1) and CCL17 (TARC) are serological indicators of multiple helminth infections and are driven by Schistosoma mansoni infection in humans. Trop Med Int Health 18:750–760. doi: 10.1111/tmi.12095 [DOI] [PubMed] [Google Scholar]
  • 132. Cota GF, Gomes LI, Pinto BF, Santos-Oliveira JR, Da-Cruz AM, Pedrosa MS, Tafuri WL, Rabello A. 2012. Dyarrheal syndrome in a patient co-infected with Leishmania Infantum and Schistosoma mansoni. Case Rep Med 2012:240512. doi: 10.1155/2012/240512 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Gregorio AW, Vasconcellos MRA, Enokihara M, Guerra JM, Nonogaki S, Tomimori J. 2019. Cutaneous schistosomiasis and leishmaniasis coinfection: a case report. J Eur Acad Dermatol Venereol 33:1781–1783. doi: 10.1111/jdv.15521 [DOI] [PubMed] [Google Scholar]
  • 134. La Flamme AC, Scott P, Pearce EJ. 2002. Schistosomiasis delays lesion resolution during Leishmania major infection by impairing parasite killing by macrophages. Parasite Immunol 24:339–345. doi: 10.1046/j.1365-3024.2002.00473.x [DOI] [PubMed] [Google Scholar]
  • 135. Hassan MF, Zhang Y, Engwerda CR, Kaye PM, Sharp H, Bickle QD. 2006. The Schistosoma mansoni hepatic egg granuloma provides a favorable microenvironment for sustained growth of Leishmania donovani. Am J Pathol 169:943–953. doi: 10.2353/ajpath.2006.051319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Khayeka-Wandabwa C, Kutima H, Nyambati VCS, Ingonga J, Oyoo-Okoth E, Karani L, Jumba B, Githuku K, Anjili CO. 2013. Combination therapy using Pentostam and Praziquantel improves lesion healing and parasite resolution in BALB/c mice co-infected with Leishmania major and Schistosoma mansoni. Parasit Vectors 6:244. doi: 10.1186/1756-3305-6-244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Camelo GMA, Silva J de O, Geiger SM, Melo MN, Negrão-Corrêa DA. 2023. Schistosoma and Leishmania: an untold story of coinfection. TropicalMed 8:383. doi: 10.3390/tropicalmed8080383 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Neglected Tropical Diseases. https://www.who.int/health-topics/neglected-tropical-diseases#tab=tab_1.
  • 139. Franco JR, Cecchi G, Paone M, Diarra A, Grout L, Kadima Ebeja A, Simarro PP, Zhao W, Argaw D. 2022. The elimination of human African trypanosomiasis: achievements in relation to WHO road map targets for 2020. PLoS Negl Trop Dis 16:e0010047. doi: 10.1371/journal.pntd.0010047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Auty H, Morrison LJ, Torr SJ, Lord J. 2016. Transmission dynamics of rhodesian sleeping sickness at the interface of wildlife and livestock areas. Trends Parasitol 32:608–621. doi: 10.1016/j.pt.2016.05.003 [DOI] [PubMed] [Google Scholar]
  • 141.Chagas disease (also known as American trypanosomiasis). https://www.who.int/news-room/fact-sheets/detail/chagas-disease-(american-trypanosomiasis).
  • 142. Chagas disease - PAHO/WHO | Pan American Health Organization. https://www.paho.org/en/topics/chagas-disease.
  • 143. Simarro PP, Cecchi G, Franco JR, Paone M, Diarra A, Ruiz-Postigo JA, Fèvre EM, Mattioli RC, Jannin JG. 2012. Estimating and mapping the population at risk of sleeping sickness. PLoS Negl Trop Dis 6:e1859. doi: 10.1371/journal.pntd.0001859 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Kloetzel K, Faleiros JJ, Mendes SR, Stanley CT, Sanches Arias H. 1973. Concomitant infection of albino mice by Trypanosoma cruzi and Schistosoma mansoni. Parasitological parameters. Trans R Soc Trop Med Hyg 67:652–658. doi: 10.1016/0035-9203(73)90034-5 [DOI] [PubMed] [Google Scholar]
  • 145. Rodrigues JPF, Caldas IS, Gonçalves RV, Almeida LA, Souza RLM, Novaes RD. 2017. S. mansoni-T. cruzi co-infection modulates arginase-1/iNOS expression, liver and heart disease in mice. Nitric Oxide 66:43–52. doi: 10.1016/j.niox.2017.02.013 [DOI] [PubMed] [Google Scholar]
  • 146. Hams E, Aviello G, Fallon PG. 2013. The Schistosoma granuloma: friend or foe?. Front Immunol 4:89. doi: 10.3389/fimmu.2013.00089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Dunne DW, Lucas S, Bickle Q, Pearson S, Madgwick L, Bain J, Doenhoff MJ. 1981. Identification and partial purification of an antigen (ω1) from Schistosoma mansoni eggs which is putatively hepatotoxic in T-cell deprived mice. Trans R Soc Trop Med Hyg 75:54–71. doi: 10.1016/0035-9203(81)90013-4 [DOI] [PubMed] [Google Scholar]
  • 148. Fagbemi BO. 1987. Interference with homologous immunity and faecal egg excretion in Schistosoma infections in mice concurrently infected with Trypanosoma brucei and Schistosoma mansoni. Folia Parasitol (Praha) 34:317–322. [PubMed] [Google Scholar]
  • 149. Fagbemi BO, Christensen NO, Dipeolu OO. 1987. Effects of Trypanosoma brucei and Babesia microti infections on the primary granulomatous reaction to Schistosoma eggs in mice. Lab Anim 21:121–124. doi: 10.1177/002367728702100207 [DOI] [PubMed] [Google Scholar]
  • 150. Genaro O, Brener Z, Coelho PM. 1986. Schistosoma mansoni: immunodepression of hepatic schistosome granuloma formation in mice infected by Trypanosoma cruzi. Rev Soc Bras Med Trop 19:35–37. doi: 10.1590/s0037-86821986000100008 [DOI] [PubMed] [Google Scholar]
  • 151. Prevention . 2019. C.-C. for D. C. and. CDC - Toxoplasmosis - Epidemiology & Risk Factors
  • 152. de-la-Torre A, Sauer A, Pfaff AW, Bourcier T, Brunet J, Speeg-Schatz C, Ballonzoli L, Villard O, Ajzenberg D, Sundar N, Grigg ME, Gomez-Marin JE, Candolfi E. 2013. Severe South American ocular toxoplasmosis is associated with decreased Ifn-γ/Il-17a and increased Il-6/Il-13 intraocular levels. PLoS Negl Trop Dis 7:e2541. doi: 10.1371/journal.pntd.0002541 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Pernas L, Ramirez R, Holmes TH, Montoya JG, Boothroyd JC. 2014. Immune profiling of pregnant Toxoplasma-infected US and Colombia patients reveals surprising impacts of infection on peripheral blood cytokines. J Infect Dis 210:923–931. doi: 10.1093/infdis/jiu189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. de la Mata Navazo S, Slöcker Barrio M, García-Morín M, Beléndez C, Escobar Fernández L, Rincón-López EM, Aguilera Alonso D, Guinea J, Marín M, Butragueño-Laiseca L, López-Herce Cid J. 2021. Case report: severe ARDS in a pediatric hematopoietic stem-cell transplantation recipient caused by disseminated toxoplasmosis. Front Pediatr 9:810718. doi: 10.3389/fped.2021.810718 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Montoya JG, Liesenfeld O. 2004. Toxoplasmosis. Lancet 363:1965–1976. doi: 10.1016/S0140-6736(04)16412-X [DOI] [PubMed] [Google Scholar]
  • 156. Figueroa-Vercellino JP, Miguel L, Moll-Udina A, Alba-Linero C, Llorenç V, Adán A. 2021. Atypical toxoplasmic retinochoroiditis in patients with malignant hematological diseases. Arch Soc Esp Oftalmol (Engl Ed) 96:152–156. doi: 10.1016/j.oftal.2020.05.032 [DOI] [PubMed] [Google Scholar]
  • 157. Dubey JP, Murata FHA, Cerqueira-Cézar CK, Kwok OCH, Villena I. 2021. Congenital toxoplasmosis in humans: an update of worldwide rate of congenital infections. Parasitology 148:1406–1416. doi: 10.1017/S0031182021001013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Koblansky AA, Jankovic D, Oh H, Hieny S, Sungnak W, Mathur R, Hayden MS, Akira S, Sher A, Ghosh S. 2013. Recognition of profilin by Toll-like receptor 12 is critical for host resistance to Toxoplasma gondii. Immunity 38:119–130. doi: 10.1016/j.immuni.2012.09.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Plattner F, Yarovinsky F, Romero S, Didry D, Carlier M-F, Sher A, Soldati-Favre D. 2008. Toxoplasma profilin is essential for host cell invasion and TLR11-dependent induction of an interleukin-12 response. Cell Host Microbe 3:77–87. doi: 10.1016/j.chom.2008.01.001 [DOI] [PubMed] [Google Scholar]
  • 160. Johnson LL, Sayles PC. 2002. Deficient humoral responses underlie susceptibility to Toxoplasma gondii in CD4-deficient mice. Infect Immun 70:185–191. doi: 10.1128/IAI.70.1.185-191.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Deshmukh AS, Gurupwar R, Mitra P, Aswale K, Shinde S, Chaudhari S. 2021. Toxoplasma gondii induces robust humoral immune response against cyst wall antigens in chronically infected animals and humans. Microb Pathog 152:104643. doi: 10.1016/j.micpath.2020.104643 [DOI] [PubMed] [Google Scholar]
  • 162. Suzuki Y, Orellana MA, Schreiber RD, Remington JS. 1988. Interferon-gamma: the major mediator of resistance against Toxoplasma gondii. Science 240:516–518. doi: 10.1126/science.3128869 [DOI] [PubMed] [Google Scholar]
  • 163. Bigna JJ, Tochie JN, Tounouga DN, Bekolo AO, Ymele NS, Youda EL, Sime PS, Nansseu JR. 2020. Global, regional, and country seroprevalence of Toxoplasma gondii in pregnant women: a systematic review, modelling and meta-analysis. Sci Rep 10:12102. doi: 10.1038/s41598-020-69078-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Khan MB, Sonaimuthu P, Lau YL, Al-Mekhlafi HM, Mahmud R, Kavana N, Kassuku A, Kasanga C. 2014. High seroprevalence of echinococossis, schistosomiasis and toxoplasmosis among the populations in Babati and Monduli districts, Tanzania. Parasit Vectors 7:505. doi: 10.1186/s13071-014-0505-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Felician EK, Ngoda OA, Jahanpour OF, Kahima J, Msuya SE, Lukambagire AH. 2022. Placental parasitic infections and pregnancy outcomes among women delivering at a tertiary hospital in northern Tanzania. East Afr Health Res J 6:141–146. doi: 10.24248/eahrj.v6i2.695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Afifi MA, Tawfeek GM, Abdel Aziz SS, Abdel Aaty HA. 2000. Assessment of the role of soluble intracellular adhesion molecule-1 (sICAM-1) in the pathogenesis and as a marker of disease severity in different stages of human schistosomiasis and toxoplasmosis. J Egypt Soc Parasitol 30:537–545. [PubMed] [Google Scholar]
  • 167. Fayad ME, Farrag AM, Hussein MM, Amer NA. 1992. Studies on chronic liver diseases in patients with and without Toxoplasma latent infection. J Egypt Soc Parasitol 22:807–815. [PubMed] [Google Scholar]
  • 168. Liu X, Wu M, Liu Y, Li J, Yang D, Jiang L. 2021. Foodborne parasites dominate current parasitic infections in Hunan province, China. Front Cell Infect Microbiol 11:774980. doi: 10.3389/fcimb.2021.774980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Marshall AJ, Brunet LR, van Gessel Y, Alcaraz A, Bliss SK, Pearce EJ, Denkers EY. 1999. Toxoplasma gondii and Schistosoma mansoni synergize to promote hepatocyte dysfunction associated with high levels of plasma TNF-α and early death in C57Bl/6 mice. J Immunol 163:2089–2097. [PubMed] [Google Scholar]
  • 170. Kloetzel K, Chieffi PP, Faleiros JJ, Filho TJ. 1977. Mortality and other parameters of concomitant infections in albino mice: the Schistosoma-Toxoplasma model. Trop Geogr Med 29:407–410. [PubMed] [Google Scholar]
  • 171. Pêgo B, Martinusso CA, Bernardazzi C, Ribeiro BE, de Araujo Cunha AF, de Souza Mesquita J, Nanini HF, Machado MP, Castelo-Branco MTL, Cavalcanti MG, de Souza HSP. 2019. Schistosoma mansoni coinfection attenuates murine Toxoplasma gondii-induced Crohn’s-like Ileitis by preserving the epithelial barrier and downregulating the inflammatory response. Front Immunol 10:442. doi: 10.3389/fimmu.2019.00442 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Araujo MI, Bliss SK, Suzuki Y, Alcaraz A, Denkers EY, Pearce EJ. 2001. Interleukin-12 promotes pathologic liver changes and death in mice coinfected with Schistosoma mansoni and Toxoplasma gondii. Infect Immun 69:1454–1462. doi: 10.1128/IAI.69.3.1454-1462.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Davis AN, Haque R, Petri WA. 2002. Update on protozoan parasites of the intestine. Curr Opin Gastroenterol 18:10–14. doi: 10.1097/00001574-200201000-00003 [DOI] [PubMed] [Google Scholar]
  • 174. Berhe B, Bugssa G, Bayisa S, Alemu M. 2018. Foodborne intestinal protozoan infection and associated factors among patients with watery diarrhea in Northern Ethiopia; a cross-sectional study. J Health Popul Nutr 37:5. doi: 10.1186/s41043-018-0137-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Fitri LE, Candradikusuma D, Setia YD, Wibawa PA, Iskandar A, Winaris N, Pawestri AR. 2022. Diagnostic methods of common intestinal protozoa: current and future immunological and molecular methods. Trop Med Infect Dis 7:253. doi: 10.3390/tropicalmed7100253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. CDC - DPDx - Amebiasis. https://www.cdc.gov/dpdx/amebiasis/index.html.
  • 177. CDC - DPDx - Giardiasis. https://www.cdc.gov/dpdx/giardiasis/index.html.
  • 178. CDC - DPDx - Cryptosporidiosis. https://www.cdc.gov/dpdx/cryptosporidiosis/index.html.
  • 179. Ahmadpour E, Safarpour H, Xiao L, Zarean M, Hatam-Nahavandi K, Barac A, Picot S, Rahimi MT, Rubino S, Mahami-Oskouei M, Spotin A, Nami S, Baghi HB. 2020. Cryptosporidiosis in HIV-positive patients and related risk factors: a systematic review and meta-analysis. Parasite 27:27. doi: 10.1051/parasite/2020025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Squire SA, Ryan U. 2017. Cryptosporidium and Giardia in Africa: current and future challenges. Parasit Vectors 10:195. doi: 10.1186/s13071-017-2111-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Kantor M, Abrantes A, Estevez A, Schiller A, Torrent J, Gascon J, Hernandez R, Ochner C. 2018. Entamoeba histolytica: updates in clinical manifestation, pathogenesis, and vaccine development. Can J Gastroenterol Hepatol 2018:4601420. doi: 10.1155/2018/4601420 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Haque R, Huston CD, Hughes M, Houpt E, Petri WA. 2003. Amebiasis. N Engl J Med 348:1565–1573. doi: 10.1056/NEJMra022710 [DOI] [PubMed] [Google Scholar]
  • 183. Sardinha-Silva A, Alves-Ferreira EVC, Grigg ME. 2022. Intestinal immune responses to commensal and pathogenic protozoa. Front Immunol 13:963723. doi: 10.3389/fimmu.2022.963723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Gurung P, Kanneganti TD. 2016. Immune responses against protozoan parasites: a focus on the emerging role of Nod-like receptors. Cell Mol Life Sci 73:3035–3051. doi: 10.1007/s00018-016-2212-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Nakada-Tsukui K, Nozaki T. 2016. Immune response of amebiasis and immune evasion by Entamoeba histolytica. Front Immunol 7:175. doi: 10.3389/fimmu.2016.00175 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Lechner CJ, Komander K, Hegewald J, Huang X, Gantin RG, Soboslay PT, Agossou A, Banla M, Köhler C. 2013. Cytokine and chemokine responses to helminth and protozoan parasites and to fungus and mite allergens in neonates, children, adults, and the elderly. Immun Ageing 10:29. doi: 10.1186/1742-4933-10-29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Hegewald J, Gantin RG, Lechner CJ, Huang X, Agosssou A, Agbeko YF, Soboslay PT, Köhler C. 2015. Cellular cytokine and chemokine responses to parasite antigens and fungus and mite allergens in children co-infected with helminthes and protozoa parasites. J Inflamm (Lond) 12:5. doi: 10.1186/s12950-015-0050-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Al-Shehri H, Stanton MC, LaCourse JE, Atuhaire A, Arinaitwe M, Wamboko A, Adriko M, Kabatereine NB, Stothard JR. 2016. An extensive burden of giardiasis associated with intestinal schistosomiasis and anaemia in school children on the shoreline of Lake Albert, Uganda. Trans R Soc Trop Med Hyg 110:597–603. doi: 10.1093/trstmh/trw072 [DOI] [PubMed] [Google Scholar]
  • 189. Weerakoon KG, Gordon CA, Williams GM, Cai P, Gobert GN, Olveda RM, Ross AG, Olveda DU, McManus DP. 2018. Co-parasitism of intestinal protozoa and Schistosoma japonicum in a rural community in the Philippines. Infect Dis Poverty 7:121. doi: 10.1186/s40249-018-0504-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Ketema W, Taye K, Tagesse N, Shibeshi MS, Alemayehu B, G/Tsadik F, Girma B, Teklehaymanote A, Debiso A. 2022. Fulminant hepatitis and ulcerative colitis: case report of Ethiopian child with schistosomiasis and amebiasis co-infection. Int Med Case Rep J 15:409–418. doi: 10.2147/IMCRJ.S377632 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Archer J, O’Halloran L, Al-Shehri H, Summers S, Bhattacharyya T, Kabaterine NB, Atuhaire A, Adriko M, Arianaitwe M, Stewart M, LaCourse EJ, Webster BL, Bustinduy AL, Stothard JR. 2020. Intestinal schistosomiasis and giardiasis co-infection in sub-Saharan Africa: can a one health approach improve control of each waterborne parasite simultaneously. Trop Med Infect Dis 5:137. doi: 10.3390/tropicalmed5030137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. McCreesh N, Mohlamonyane M, Edwards A, Olivier S, Dikgale K, Dayi N, Gareta D, Wood R, Grant AD, White RG, Middelkoop K. 2022. Improving estimates of social contact patterns for airborne transmission of respiratory pathogens. Emerg Infect Dis 28:2016–2026. doi: 10.3201/eid2810.212567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Tuberculosis. https://www.who.int/health-topics/tuberculosis#tab=tab_1.
  • 194. Lin PL, Rutledge T, Green AM, Bigbee M, Fuhrman C, Klein E, Flynn JL. 2012. CD4 T cell depletion exacerbates acute Mycobacterium tuberculosis while reactivation of latent infection is dependent on severity of tissue depletion in cynomolgus macaques. AIDS Res Hum Retroviruses 28:1693–1702. doi: 10.1089/AID.2012.0028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, Orme IM. 1993. Disseminated tuberculosis in interferon gamma gene-disrupted mice. J Exp Med 178:2243–2247. doi: 10.1084/jem.178.6.2243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Mhimbira F, Hella J, Said K, Kamwela L, Sasamalo M, Maroa T, Chiryamkubi M, Mhalu G, Schindler C, Reither K, Knopp S, Utzinger J, Gagneux S, Fenner L, Hsieh MH. 2017. Prevalence and clinical relevance of helminth co-infections among tuberculosis patients in urban Tanzania. PLoS Negl Trop Dis 11:e0005342. doi: 10.1371/journal.pntd.0005342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Sikalengo G, Hella J, Mhimbira F, Rutaihwa LK, Bani F, Ndege R, Sasamalo M, Kamwela L, Said K, Mhalu G, Mlacha Y, Hatz C, Knopp S, Gagneux S, Reither K, Utzinger J, Tanner M, Letang E, Weisser M, Fenner L. 2018. Distinct clinical characteristics and helminth co-infections in adult tuberculosis patients from urban compared to rural Tanzania. Infect Dis Poverty 7:24. doi: 10.1186/s40249-018-0404-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Bettencourt PJG, Joosten SA, Lindestam Arlehamn CS, Behr MA, Locht C, Neyrolles O. 2021. 100 years of the Bacillus Calmette-Guérin vaccine. Vaccine 39:7221–7222. doi: 10.1016/j.vaccine.2021.11.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Elias D, Akuffo H, Thors C, Pawlowski A, Britton S. 2005. Low dose chronic Schistosoma mansoni infection increases susceptibility to Mycobacterium bovis BCG infection in mice. Clin Exp Immunol 139:398–404. doi: 10.1111/j.1365-2249.2004.02719.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Elias D, Akuffo H, Pawlowski A, Haile M, Schön T, Britton S. 2005. Schistosoma mansoni infection reduces the protective efficacy of BCG vaccination against virulent Mycobacterium tuberculosis. Vaccine 23:1326–1334. doi: 10.1016/j.vaccine.2004.09.038 [DOI] [PubMed] [Google Scholar]
  • 201. Gopal R, Monin L, Torres D, Slight S, Mehra S, McKenna KC, Fallert Junecko BA, Reinhart TA, Kolls J, Báez-Saldaña R, et al. 2013. S100A8/A9 proteins mediate neutrophilic inflammation and lung pathology during tuberculosis. Am J Respir Crit Care Med 188:1137–1146. doi: 10.1164/rccm.201304-0803OC [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Slight SR, Rangel-Moreno J, Gopal R, Lin Y, Fallert Junecko BA, Mehra S, Selman M, Becerril-Villanueva E, Baquera-Heredia J, Pavon L, Kaushal D, Reinhart TA, Randall TD, Khader SA. 2013. CXCR5+ T helper cells mediate protective immunity against tuberculosis. J Clin Invest 123:712–726. doi: 10.1172/JCI65728 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Monin L, Griffiths KL, Lam WY, Gopal R, Kang DD, Ahmed M, Rajamanickam A, Cruz-Lagunas A, Zúñiga J, Babu S, Kolls JK, Mitreva M, Rosa BA, Ramos-Payan R, Morrison TE, Murray PJ, Rangel-Moreno J, Pearce EJ, Khader SA. 2015. Helminth-induced arginase-1 exacerbates lung inflammation and disease severity in tuberculosis. J Clin Invest 125:4699–4713. doi: 10.1172/JCI77378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Munder M. 2009. Arginase: an emerging key player in the mammalian immune system. Br J Pharmacol 158:638–651. doi: 10.1111/j.1476-5381.2009.00291.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. El-Gayar S, Thüring-Nahler H, Pfeilschifter J, Röllinghoff M, Bogdan C. 2003. Helminth-induced arginase-1 exacerbates lung inflammation and disease severity in tuberculosis. J Immunol 171:4561–4568. doi: 10.4049/jimmunol.171.9.4561 [DOI] [PubMed] [Google Scholar]
  • 206. Toulza F, Tsang L, Ottenhoff THM, Brown M, Dockrell HM. 2016. Mycobacterium tuberculosis-specific CD4+ T-cell response is increased, and treg cells decreased, in anthelmintic-treated patients with latent TB. Eur J Immunol 46:752–761. doi: 10.1002/eji.201545843 [DOI] [PubMed] [Google Scholar]
  • 207. Sharma SK, Mohan A, Kadhiravan T. 2005. HIV-TB co-infection: epidemiology, diagnosis & management. Indian J Med Res 121:550–567. [PubMed] [Google Scholar]
  • 208. DiNardo AR, Mace EM, Lesteberg K, Cirillo JD, Mandalakas AM, Graviss EA, Orange JS, Makedonas G. 2016. Schistosome soluble egg antigen decreases Mycobacterium tuberculosis–specific CD4+ T-cell effector function with concomitant arrest of macrophage phago-lysosome maturation. J Infect Dis 214:479–488. doi: 10.1093/infdis/jiw156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. DiNardo AR, Nishiguchi T, Mace EM, Rajapakshe K, Mtetwa G, Kay A, Maphalala G, Secor WE, Mejia R, Orange JS, Coarfa C, Bhalla KN, Graviss EA, Mandalakas AM, Makedonas G. 2018. Schistosomiasis induces persistent DNA methylation and tuberculosis-specific immune changes. J Immunol 201:124–133. doi: 10.4049/jimmunol.1800101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Harari A, Rozot V, Bellutti Enders F, Perreau M, Stalder JM, Nicod LP, Cavassini M, Calandra T, Blanchet CL, Jaton K, Faouzi M, Day CL, Hanekom WA, Bart P-A, Pantaleo G. 2011. Dominant TNF-α+ Mycobacterium tuberculosis-specific CD4+ T cell responses discriminate between latent infection and active disease. Nat Med 17:372–376. doi: 10.1038/nm.2299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Potian JA, Rafi W, Bhatt K, McBride A, Gause WC, Salgame P. 2011. Preexisting helminth infection induces inhibition of innate pulmonary anti-tuberculosis defense by engaging the IL-4 receptor pathway. J Exp Med 208:1863–1874. doi: 10.1084/jem.20091473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Schick J, Altunay M, Lacorcia M, Marschner N, Westermann S, Schluckebier J, Schubart C, Bodendorfer B, Christensen D, Alexander C, Wirtz S, Voehringer D, da Costa CP, Lang R. 2023. IL-4 and helminth infection downregulate MINCLE-dependent macrophage response to Mycobacteria and Th17 adjuvanticity. Elife 12:e72923. doi: 10.7554/eLife.72923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Togarsimalemath SK, Pushpamithran G, Schön T, Stendahl O, Blomgran R. 2021. Helminth antigen exposure enhances early immune control of Mycobacterium tuberculosis in monocytes and macrophages. J Innate Immun 13:148–163. doi: 10.1159/000512279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Aira N, Andersson AM, Singh SK, McKay DM, Blomgran R. 2017. Species dependent impact of helminth-derived antigens on human macrophages infected with Mycobacterium tuberculosis: direct effect on the innate anti-mycobacterial response. PLoS Negl Trop Dis 11:e0005390. doi: 10.1371/journal.pntd.0005390 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. McLaughlin TA, Khayumbi J, Ongalo J, Tonui J, Campbell A, Allana S, Gurrion Ouma S, Odhiambo FH, Gandhi NR, Day CL. 2020. CD4 T cells in Mycobacterium tuberculosis and Schistosoma mansoni co-infected individuals maintain functional Th1 responses. Front Immunol 11:127. doi: 10.3389/fimmu.2020.00127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Bewket G, Kiflie A, Abate E, Stendahl O, Schön T, Blomgran R. 2021. Helminth species specific expansion and increased TNF-alpha production of non-classical monocytes during active tuberculosis. PLoS Negl Trop Dis 15:e0009194. doi: 10.1371/journal.pntd.0009194 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Kiflie A, Bewket G, Abate E, Schön T, Blomgran R. 2021. Differential effects of asymptomatic Ascaris lumbricoides, Schistosoma mansoni or hook worm infection on the frequency and TGF-beta-producing capacity of regulatory T cells during active tuberculosis. Tuberculosis (Edinb) 131:102126. doi: 10.1016/j.tube.2021.102126 [DOI] [PubMed] [Google Scholar]
  • 218. McLaughlin TA, Khayumbi J, Ongalo J, Matete D, Tonui J, Muchiri B, Sasser LE, Campbell A, Allana S, Ouma SG, Hayara FO, Gandhi NR, Day CL. 2020. Adults from Kisumu, Kenya have robust γδ T cell responses to Schistosoma mansoni, which are modulated by tuberculosis. PLoS Negl Trop Dis 14:e0008764. doi: 10.1371/journal.pntd.0008764 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Dennison CL, de Oliveira LB, Fraga LA de O, E Lima RS, Ferreira JA, Clennon JA, de Mondesert L, Stephens J, Magueta EB, Castelo Branco A, Rezende M de C, Negrão-Corrêa D, de Faria Grossi MA, Fairley JK. 2021. Mycobacterium leprae-helminth co-infections and vitamin D deficiency as potential risk factors for leprosy: a case-control study in south-eastern Brazil. Int J Infect Dis 105:261–266. doi: 10.1016/j.ijid.2021.02.048 [DOI] [PubMed] [Google Scholar]
  • 220. Phillips DA, Ferreira JA, Ansah D, Teixeira HS, Kitron U, Filippis T de, Alcântara M de, Fairley JK. 2017. A tale of two neglected tropical infections: using GIS to assess the spatial and temporal overlap of schistosomiasis and leprosy in a region of Minas Gerais, Brazil. Mem Inst Oswaldo Cruz 112:275–280. doi: 10.1590/0074-02760160395 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Emerson LE, Anantharam P, Yehuala FM, Bilcha KD, Tesfaye AB, Fairley JK. 2020. Poor WASH (water, sanitation, and hygiene) conditions are associated with leprosy in North Gondar, Ethiopia. Int J Environ Res Public Health 17:1–10. doi: 10.3390/ijerph17176061 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222. 2015. WHO Estimates of the Global Burden of Foodborne Diseases: Foodborne Disease Burden Epidemiology Reference Group 2007-2015. Available from: https://www.who.int/publications/i/item/9789241565165
  • 223. Salmonella Homepage | CDC. n.d. Available from: https://www.cdc.gov/salmonella/index.html
  • 224. Fierer J. 2022. Invasive non-typhoidal Salmonella (iNTS) infections. Clin Infect Dis 75:732–738. doi: 10.1093/cid/ciac035 [DOI] [PubMed] [Google Scholar]
  • 225. Dandekar T, Astrid F, Jasmin P, Hensel M. 2012. Salmonella enterica: a surprisingly well-adapted intracellular lifestyle. Front Microbiol 3:164. doi: 10.3389/fmicb.2012.00164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Eng S-K, Pusparajah P, Ab Mutalib N-S, Ser H-L, Chan K-G, Lee L-H. 2015. Salmonella: a review on pathogenesis, epidemiology and antibiotic resistance. Frontiers in Life Science 8:284–293. doi: 10.1080/21553769.2015.1051243 [DOI] [Google Scholar]
  • 227. Darby J, Sheorey H. 2008. Searching for Salmonella. Aust Fam Physician 37:806–810. [PubMed] [Google Scholar]
  • 228. Trawinski H, Wendt S, Lippmann N, Heinitz S, von Braun A, Lübbert C. 2020. Typhoid and Paratyphoid fever. Z Gastroenterol 58:160–170. doi: 10.1055/a-1063-1945 [DOI] [PubMed] [Google Scholar]
  • 229. Information for Healthcare Professionals | Salmonella | CDC. https://www.cdc.gov/salmonella/general/technical.html.
  • 230. Mastroeni P, Rossi O. 2020. Antibodies and protection in systemic Salmonella infections: do we still have more questions than answers Infect Immun 88:e00219-20. doi: 10.1128/IAI.00219-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Huang FC. 2021. The interleukins orchestrate mucosal immune responses to Salmonella infection in the intestine. Cells 10:3492. doi: 10.3390/cells10123492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Ingram JP, Brodsky IE, Balachandran S. 2017. Interferon–γ in Salmonella pathogenesis: new tricks for an old dog. Cytokine 98:27–32. doi: 10.1016/j.cyto.2016.10.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233. Rogers AWL, Tsolis RM, Bäumler AJ. 2020. Salmonella versus the microbiome. Microbiol Mol Biol Rev 85. doi: 10.1128/MMBR.00027-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234. Bajinka O, Qi M, Barrow A, Touray AO, Yang L, Tan Y. 2021. Pathogenicity of Salmonella during Schistosoma-Salmonella co-infections and the importance of the gut microbiota. Curr Microbiol 79:26. doi: 10.1007/s00284-021-02718-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. Muniz-Junqueira MI, Tosta CE, Prata A. 2009. Schistosoma-associated chronic septicemic salmonellosis: evolution of knowledge and immunopathogenic mechanisms. Rev Soc Bras Med Trop 42:436–445. doi: 10.1590/s0037-86822009000400015 [DOI] [PubMed] [Google Scholar]
  • 236. Bourée P, Botterel F, Romand S. 2002. Delayed Salmonella bacteriuria in a patient infected with Schistosoma haematobium. J Egypt Soc Parasitol 32:355–360. [PubMed] [Google Scholar]
  • 237. Strausbaugh LJ, Farid Z, Higashi GI, Bassily S. 1978. Renal amyloidosis with nephrotic syndrome in two patients with Schistosomiasis mansoni and chronic salmonellosis. Am J Trop Med Hyg 27:919–923. doi: 10.4269/ajtmh.1978.27.919 [DOI] [PubMed] [Google Scholar]
  • 238. Marege A, Seid M, Boke B, Thomas S, Arage M, Mouze N, Yohanes T, Woldemariam M, Manilal A. 2021. Prevalence of Schistosoma mansoni-Salmonella coinfection among patients in southern Ethiopia. New Microbes and New Infections 40:100842. doi: 10.1016/j.nmni.2021.100842 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Gendrel D, Kombila M, Beaudoin-Leblevec G, Richard-Lenoble D. 1994. Nontyphoidal salmonellal septicemia in gabonese children infected with Schistosoma intercalatum. Clin Infect Dis 18:103–105. doi: 10.1093/clinids/18.1.103 [DOI] [PubMed] [Google Scholar]
  • 240. Mbuyi-Kalonji L, Barbé B, Nkoji G, Madinga J, Roucher C, Linsuke S, Hermy M, Heroes A-S, Mattheus W, Polman K, Lutumba P, Phoba M-F, Lunguya O, Jacobs J. 2020. Non-typhoidal salmonella intestinal carriage in a Schistosoma mansoni endemic community in a rural area of the democratic republic of congo. PLoS Negl Trop Dis 14:e0007875. doi: 10.1371/journal.pntd.0007875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241. Chacha F, Mshana SE, Mirambo MM, Mushi MF, Kabymera R, Gerwing L, Schneiderhan W, Zimmermann O, Groß U. 2015. Salmonella Typhi meningitis in a 9-year old boy with urinary schistosomiasis: a case report. BMC Res Notes 8:64. doi: 10.1186/s13104-015-1030-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242. Abdul-Fattah MM, Yossef SM, Ebraheem ME, Nasr ME, Hassan MA, Abdul Wahab SE. 1995. Schistosomal glomerulopathy: a putative role for commonly associated Salmonella infection. J Egypt Soc Parasitol 25:165–173. [PubMed] [Google Scholar]
  • 243. Martinelli R, Pereira LJ, Brito E, Rocha H. 1992. Renal involvement in prolonged Salmonella bacteremia: the role of schistosomal glomerulopathy. Rev Inst Med Trop Sao Paulo 34:193–198. doi: 10.1590/s0036-46651992000300002 [DOI] [PubMed] [Google Scholar]
  • 244. Ashour DS, Othman AA. 2020. Parasite–bacteria interrelationship. Parasitol Res 119:3145–3164. doi: 10.1007/s00436-020-06804-2 [DOI] [PubMed] [Google Scholar]
  • 245. Melhem RF, LoVerde PT. 1984. Mechanism of interaction of Salmonella and Schistosoma mansoni species. Infect Immun 44:274–281. doi: 10.1128/iai.44.2.274-281.1984 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Barnhill AE, Novozhilova E, Day TA, Carlson SA. 2011. Schistosoma mansoni-associated Salmonella resist antibiotics via specific fimbrial attachments to the flatworm. Parasit Vectors 4:123. doi: 10.1186/1756-3305-4-123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247. Njunda AL, Oyerinde JP. 1996. Salmonella typhi infection in Schistosoma mansoni infected mice. West Afr J Med 15:24–30. [PubMed] [Google Scholar]
  • 248. Schramm G, Suwandi A, Galeev A, Sharma S, Braun J, Claes A-K, Braubach P, Grassl GA. 2018. Schistosome eggs impair protective Th1/Th17 immune responses against Salmonella infection. Front Immunol 9:2614. doi: 10.3389/fimmu.2018.02614 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. Lam HYP, Wu W-J, Liang T-R, Li H-C, Chang K-C, Peng S-Y. 2023. Salmonella typhimurium exacerbates injuries but resolves fibrosis in liver and spleen during Schistosoma mansoni infection. J Microbiol Immunol Infect 56:477–489. doi: 10.1016/j.jmii.2023.03.002 [DOI] [PubMed] [Google Scholar]
  • 250. Muniz-Junqueira MI, Prata A, Tosta CE. 1992. Phagocytic and bactericidal function of mouse macrophages to Salmonella typhimurium in Schistosoma mansoni. Am J Trop Med Hyg 46:132–136. doi: 10.4269/ajtmh.1992.46.132 [DOI] [PubMed] [Google Scholar]
  • 251. Muniz-Junqueira MI, Prata A, Tosta CE. 1997. Factors influencing phagocytosis of Salmonella typhimurium by macrophages in murine schistosomiasis. Rev Soc Bras Med Trop 30:101–106. doi: 10.1590/s0037-86821997000200003 [DOI] [PubMed] [Google Scholar]
  • 252. Mikhail IA, Higashi GI, Mansour NS, Edman DC, Elwan SH. 1981. Salmonella paratyphi A in hamsters concurrently infected with Schistosoma mansoni. Am J Trop Med Hyg 30:385–393. doi: 10.4269/ajtmh.1981.30.385 [DOI] [PubMed] [Google Scholar]
  • 253. Zhu X, Chen L, Wu J, Tang H, Wang Y. 2017. Salmonella typhimurium infection reduces Schistosoma japonicum worm burden in mice. Sci Rep 7:1349. doi: 10.1038/s41598-017-00992-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Foxman B. 2002. Epidemiology of urinary tract infections: incidence, morbidity, and economic costs. Am J Med 113 Suppl 1A:5S–13S. doi: 10.1016/s0002-9343(02)01054-9 [DOI] [PubMed] [Google Scholar]
  • 255. Harrington RD, Hooton TM. 2000. Urinary tract infection risk factors and gender. J Gend Specif Med 3:27–34. [PubMed] [Google Scholar]
  • 256. Doherty JF, Grant AD, Bryceson AD. 1995. Fever as the presenting complaint of travellers returning from the tropics. QJM 88:277–281. doi: 10.1093/oxfordjournals.qjmed.a069060 [DOI] [PubMed] [Google Scholar]
  • 257. Nji CP, Assob JCN, Akoachere J-F, Yokota S. 2020. Predictors of urinary tract infections in children and antibiotic susceptibility pattern in the Buea health district. Biomed Res Int 2020:2176569. doi: 10.1155/2020/2176569 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Eyong ME, Ikepeme EE, Ekanem EE. 2008. Relationship between Schistosoma haematobium infection and urinary tract infection among children in South Eastern, Nigeria. Niger Postgrad Med J 15:89–93. [PubMed] [Google Scholar]
  • 259. Adeyeba O, Ojeaga S. 2010. Urinary schistosomiasis and concomitant urinary tract pathogens among school children in metropoitan Ibadan, Nigeria. Afr J Biomed Res 5:103–107. doi: 10.4314/ajbr.v5i3.53993 [DOI] [Google Scholar]
  • 260. Chai Z-W, Xu Q-C, Xu C-M. 2014. Schistosomiasis haematobia misdiagnosed as urinary trace infection: one case report. Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi 26:I. [PubMed] [Google Scholar]
  • 261. Jones J. 1977. Schistosomiasis haematobium. JAMA 238:1275–1276. doi: 10.1001/jama.1977.03280130057018 [DOI] [PubMed] [Google Scholar]
  • 262. Seynabou L, Awa BD, Oumarou FD, Moustapha M, Makhtar C, Mamadou D, Rokhaya D, Mamadou LD, G eacute rard CD, Roughyatou K, Th eacute r egrave se D, Babacar F, Ahmad IS. 2016. Profile of bacterial and parasitic urinary infections in Saint Louis Senegal between 2000 and 2010. Afr J Microbiol Res 10:1061–1065. doi: 10.5897/AJMR2015.7838 [DOI] [Google Scholar]
  • 263. Gomes CM, Hisano M, Machado LR, Figueiredo JA, Lucon AM, Trigo-Rocha FE. 2005. Urological manifestations of chronic schistosomal myeloradiculopathy. BJU Int 96:853–856. doi: 10.1111/j.1464-410X.2005.05726.x [DOI] [PubMed] [Google Scholar]
  • 264. el Hawey A, Massoud A, Badr el Din N, Waheeb A, Abd-el Hamid S. 1989. Bacterial flora in hepatic encephalopathy in bilharzial and non-bilharzial patients. J Egypt Soc Parasitol 19:797–804. [PubMed] [Google Scholar]
  • 265. Sobh MA, el-Agroudy AE, Moustafa FE, Shokeir AA, el-Shazly A, Ghoneim MA. 1992. Impact of schistosomiasis on patient and graft outcome after kidney transplantation. Nephrol Dial Transplant 7:858–864. [PubMed] [Google Scholar]
  • 266. Gomes CM, Trigo-Rocha F, Arap MA, Gabriel AJ, Alaor de Figueiredo J, Arap S. 2002. Schistosomal myelopathy: urologic manifestations and urodynamic findings. Urology 59:195–200. doi: 10.1016/s0090-4295(01)01508-4 [DOI] [PubMed] [Google Scholar]
  • 267. Nmorsi OPG, Kwandu UNCD, Ebiaguanye LM. 2007. Schistosoma haematobium and urinary tract pathogens co-infections in a rural community of Edo State, Nigeria. J Commun Dis 39:85–90. [PubMed] [Google Scholar]
  • 268. J.Amoo K, Amoo O, Oke A, Adegboyega T. 2017. Prevalence of urinary tract infection (UTI) and concomitant urinary schistosomiasis among primary school children in Remo North local government, Ogun state, Nigeria. IOSR Journal of Dental and Medical Sciences (IOSR-JDMS) 16:68–73. [Google Scholar]
  • 269. Ayoade F, Moro DD, Ebene OL. 2013. Prevalence and antimicrobial susceptibility pattern of asymptomatic urinary tract infections of bacterial and parasitic origins among university students in redemption camp, Ogun state, Nigeria. OJMM 03:219–226. doi: 10.4236/ojmm.2013.34033 [DOI] [Google Scholar]
  • 270. Kone KJ, Onifade AK, Dada EO. 2023. Risk factors affecting the occurrence of urinary schistosomiasis and urinary tract infections in some communities of Ondo State, Nigeria. J Water Health 21:27–34. doi: 10.2166/wh.2022.186 [DOI] [PubMed] [Google Scholar]
  • 271. Soyannwo MA, Ogbechi ME, Adeyeni GA, Soyeni AI, Lipede MR, Lucas AO. 1978. Studies on the prevalence of renal disease and hypertension in relation to schistosomiasis. III. Proteinuria, haematuria, pyuria and bacteriuria in the rural community of Nigeria. Niger Med J 8:451–464. [PubMed] [Google Scholar]
  • 272. Ossai OP, Dankoli R, Nwodo C, Tukur D, Nsubuga P, Ogbuabor D, Ekwueme O, Abonyi G, Ezeanolue E, Nguku P, Nwagbo D, Idris S, Eze G. 2014. Bacteriuria and urinary schistosomiasis in primary school children in rural communities in Enugu State, Nigeria, 2012. Pan Afr Med J 18:15. doi: 10.11604/pamj.supp.2014.18.1.4169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Chidinma Ursula Uwandu, Joy Nkeiruka Dike-Ndudim, Chizaram Winners Ndubueze 2022. Epidemiological studies on urinary schistosomiasis and bacterial co-infection in some rural communities of Abia State. World J Bio Pharm Health Sci 10:065–072. doi: 10.30574/wjbphs.2022.10.3.0079 [DOI] [Google Scholar]
  • 274. Anosike JC, et al. 2001. Endemicity of urinary schistosomiasis in north central zone of Abia state, Nigeria. Int J Environ Hlth Human Develop 1:6–15. [Google Scholar]
  • 275. Nwachukwu IO, et al. 2018. Urinary schistosomiasis and concomitant bacteriuria among school age children in some parts of Owerri, IMO state. INTERNATIONAL JOURNAL OF RESEARCH IN ADVANCE ENGINEERING 3:107–115. [Google Scholar]
  • 276. Mahmoud KM, Sobh MA, El-Agroudy AE, Mostafa FE, Baz ME, Shokeir AA, Ghoneim MA. 2001. Impact of schistosomiasis on patient and graft outcome after renal transplantation: 10 years’ follow-up. Nephrol Dial Transplant 16:2214–2221. doi: 10.1093/ndt/16.11.2214 [DOI] [PubMed] [Google Scholar]
  • 277. Wishahi MM, el-Sherbeni E, el-Baz H. 1984. Granulomatous cystitis: a reversible lesion in children. Eur Urol 10:368–369. doi: 10.1159/000463835 [DOI] [PubMed] [Google Scholar]
  • 278. Usaini B, Taura D, Koki Y, et al. 2006. Studies on seasonal variation in the occurrences of Schistosoma haematobium and bacterial urinary infections among school age children in Kano. IOSR J Pharm Biol Sci 10:27–33. [Google Scholar]
  • 279. Alagha B, Dada EO. 2021. Urinary schistosomiasis and asymptomatic bacteriuria among individuals of Ipogun, Nigeria: detection of predominant microorganisms and antibiotic susceptibility profile. jmhs 2:70–80. doi: 10.32996/jmhs.2021.2.2.8 [DOI] [Google Scholar]
  • 280. Nimorsi OP, Egwunyenga AO, Bajomo DO. 2001. Survey of urinary schistosomiasis and trichomoniasis in a rural community in Edo State, Nigeria. J Commun Dis 33:96–101. [PubMed] [Google Scholar]
  • 281. Gadoth A, Mvumbi G, Hoff NA, Musene K, Mukadi P, Ashbaugh HR, Doshi RH, Javanbakht M, Gorbach P, Okitolonda-Wemakoy E, Klausner JD, Rimoin AW. 2019. Urogenital schistosomiasis and sexually transmitted coinfections among pregnant women in a schistosome-endemic region of the democratic republic of Congo. Am J Trop Med Hyg 101:828–836. doi: 10.4269/ajtmh.19-0024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282. Hennequin C, Bourée P, Halfon P. 1991. Salmonella paratyphi A urinary infection in Schistosoma mansoni bilharziasis. Rev Med Interne 12:141–142. doi: 10.1016/s0248-8663(05)81378-5 [DOI] [PubMed] [Google Scholar]
  • 283. Uwaezuoke J, Anosike J, Udujih O, Onyeka P. 2010. Epidemiological and bacteriological studies on vesical schistosomiasis in Ikwo area, Ebonyi state, Nigeria. JASEM 12:75–80. doi: 10.4314/jasem.v12i2.55535 [DOI] [Google Scholar]
  • 284. Uneke CJ, Ugwuoke-Adibuah S, Nwakpu KO, Ngwu BA. 2009. An assessment of Schistosoma haematobium infection and urinary tract bacterial infection amongst school children in rural eastern Nigeria. IJLM 4. doi: 10.5580/270 [DOI] [Google Scholar]
  • 285. Hsieh YJ, Fu CL, Hsieh M. 2013. Interleukin-4 signaling promotes bacteriuria during urinary tract co-infection. J Urol 189:e470. doi: 10.1016/j.juro.2013.02.785 [DOI] [Google Scholar]
  • 286. Hsieh Y-J, Fu C-L, Hsieh MH, Appleton JA. 2014. Helminth-induced interleukin-4 Abrogates invariant natural killer T cell activation-associated clearance of bacterial infection. Infect Immun 82:2087–2097. doi: 10.1128/IAI.01578-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287. Mbanefo EC, Le L, Pennington LF, Hsieh Y-J, Odegaard JI, Lapira K, Jardetzky TS, Falcone FH, Hsieh MH. 2020. IPSE, a urogenital parasite-derived immunomodulatory molecule, suppresses bladder pathogenesis and anti-microbial peptide gene expression in bacterial urinary tract infection. Parasit Vectors 13:615. doi: 10.1186/s13071-020-04490-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288. Burrell CJ, Howard CR, Murphy FA. 2017. Epidemiology of viral infections. Fenner and White’s Medical Virology 185. doi: 10.1016/B978-0-12-375156-0.00013-8 [DOI] [Google Scholar]
  • 289. WHO Coronavirus (COVID-19) Dashboard | WHO Coronavirus (COVID-19) Dashboard With Vaccination Data. Available from: https://covid19.who.int
  • 290. Baker RE, Mahmud AS, Miller IF, Rajeev M, Rasambainarivo F, Rice BL, Takahashi S, Tatem AJ, Wagner CE, Wang L-F, Wesolowski A, Metcalf CJE. 2022. Infectious disease in an era of global change. Nat Rev Microbiol 20:193–205. doi: 10.1038/s41579-021-00639-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Coronavirus disease (COVID-19) pandemic. https://www.who.int/europe/emergencies/situations/covid-19.
  • 292. Feehan J, Apostolopoulos V. 2021. Is COVID-19 the worst pandemic Maturitas 149:56–58. doi: 10.1016/j.maturitas.2021.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293. Harvey WT, Carabelli AM, Jackson B, Gupta RK, Thomson EC, Harrison EM, Ludden C, Reeve R, Rambaut A, Peacock SJ, Robertson DL, COVID-19 Genomics UK (COG-UK) Consortium . 2021. SARS-CoV-2 variants, spike mutations and immune escape. Nat Rev Microbiol 19:409–424. doi: 10.1038/s41579-021-00573-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294. He X, Hong W, Pan X, Lu G, Wei X. 2021. SARS‐CoV‐2 omicron variant: characteristics and prevention. MedComm (2020) 2:838–845. doi: 10.1002/mco2.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Chang T, Jung BK, Chai JY, Cho Si. 2022. The notable global heterogeneity in the distribution of COVID-19 cases and the association with pre-existing parasitic diseases. PLoS Negl Trop Dis 16:e0010826. doi: 10.1371/journal.pntd.0010826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Nguimkeu P, Tadadjeu S. 2021. Why is the number of COVID-19 cases lower than expected in Sub-Saharan Africa? A cross-sectional analysis of the role of demographic and geographic factors. World Dev 138:105251. doi: 10.1016/j.worlddev.2020.105251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297. Favas C, Jarrett P, Ratnayake R, Watson OJ, Checchi F. 2022. Country differences in transmissibility, age distribution and case-fatality of SARS-CoV-2: a global ecological analysis. Int J Infect Dis 114:210–218. doi: 10.1016/j.ijid.2021.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298. Veldhoen M, Bertoletti A. 2023. SARS-CoV-2 clearance after breakthrough infection correlates with fit and happy T cells. Immunol Cell Biol 101:587–589. doi: 10.1111/imcb.12654 [DOI] [PubMed] [Google Scholar]
  • 299. Koutsakos M, Reynaldi A, Lee WS, Nguyen J, Amarasena T, Taiaroa G, Kinsella P, Liew KC, Tran T, Kent HE, Tan H-X, Rowntree LC, Nguyen THO, Thomas PG, Kedzierska K, Petersen J, Rossjohn J, Williamson DA, Khoury D, Davenport MP, Kent SJ, Wheatley AK, Juno JA. 2023. SARS-CoV-2 breakthrough infection induces rapid memory and de novo T cell responses. Immunity 56:879–892. doi: 10.1016/j.immuni.2023.02.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300. Bernal E, García-Villalba E, Pons E, Vicente MR, Tomás C, Minguela A, Hernández MD, Puche G, Carter P, Martinez M, Alcaraz A, Nuñez ML, Sancho N, Villalba MC, Cano A, Muñoz A, Báguena C. 2023. Role of vaccination and anti-SARS-CoV-2 antibodies in the clinical outcome of hospitalized COVID-19 patients. Med Clin (Barc) 160:476–483. doi: 10.1016/j.medcli.2022.12.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301. Bradbury RS, Piedrafita D, Greenhill A, Mahanty S. 2020. Will helminth co-infection modulate COVID-19 severity in endemic regions?. Nat Rev Immunol 20:342–342. doi: 10.1038/s41577-020-0330-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. Sánchez-Marqués R, Mas-Coma S, Salas-Coronas J, Boissier J, Bargues MD. 2022. Research on schistosomiasis in the era of the COVID-19 pandemic: a bibliometric analysis. Int J Environ Res Public Health 19:8051. doi: 10.3390/ijerph19138051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303. Wolday D, Gebrecherkos T, Arefaine ZG, Kiros YK, Gebreegzabher A, Tasew G, Abdulkader M, Abraha HE, Desta AA, Hailu A, Tollera G, Abdella S, Tesema M, Abate E, Endarge KL, Hundie TG, Miteku FK, Urban BC, Schallig H, Harris VC, de Wit TFR. 2021. Effect of co-infection with intestinal parasites on COVID-19 severity: a prospective observational cohort study. EClinicalMedicine 39:101054. doi: 10.1016/j.eclinm.2021.101054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304. Sinha P, Matthay MA, Calfee CS. 2020. Is a ‘cytokine storm’ relevant to COVID-19. JAMA Intern Med 180:1152–1154. doi: 10.1001/jamainternmed.2020.3313 [DOI] [PubMed] [Google Scholar]
  • 305. Alrouji M, Al-Kuraishy HM, Al-Gareeb AI, Elhadad H, Alexiou A, Papadakis M, Ogaly HA, Elgazzar AM, Batiha G-S. 2023. Immunological interactions in helminths-SARS CoV-2 coinfection: could old enemy be a friend today?. Parasite Immunol 45:e12982. doi: 10.1111/pim.12982 [DOI] [PubMed] [Google Scholar]
  • 306. Dantas NM, Andrade LA, Paz W da, Borges WN, Barbosa VGB, Hora D da, Silva C da, do Carmo RF, Souza C de, Santos ADD, Wanderley FS, Negrão-Corrêa DA, Fujiwara RT, Bezerra-Santos M, Porto WJN. 2023. Impact of the COVID-19 pandemic on the actions of the Schistosomiasis Control Program in an endemic area in Northeastern Brazil. Acta Trop 240:106859. doi: 10.1016/j.actatropica.2023.106859 [DOI] [PubMed] [Google Scholar]
  • 307. Li G, Xu D, Hu Y, Xu M, Zhang L, Du X, Zhang L, Sun C, Xie Y, Tan X. 2022. Impact of the coronavirus disease 2019 lockdown on Schistosoma host Oncomelania hupensis density in Wuhan. Acta Trop 226:106224. doi: 10.1016/j.actatropica.2021.106224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308. Kura K, Ayabina D, Toor J, Hollingsworth TD, Anderson RM. 2021. Disruptions to schistosomiasis programmes due to COVID-19: an analysis of potential impact and mitigation strategies. Trans R Soc Trop Med Hyg 115:236–244. doi: 10.1093/trstmh/traa202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309. About HIV/AIDS | HIV Basics | HIV/AIDS | CDC. https://www.cdc.gov/hiv/basics/whatishiv.html. [Google Scholar]
  • 310. HIV and AIDS Epidemic Global Statistics | HIV.Gov. Available from: https://www.hiv.gov/hiv-basics/overview/data-and-trends/global-statistics/
  • 311. HIV | CDC. https://www.cdc.gov/hiv/default.html.
  • 312. Gubser C, Pitman MC, Lewin SR. 2019. CD4+ T cell signatures in HIV infection. Nat. Immunol 20:948–950. [DOI] [PubMed] [Google Scholar]
  • 313. Bhattacharyya, S., Crain, C. R., Goldberg, B. & Gaiha, G. D.. Features of functional and dysfunctional CD8+ T cells to guide HIV vaccine development. Curr Opin HIV AIDS 18, 257–263 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Douek DC, Brenchley JM, Betts MR, Ambrozak DR, Hill BJ, Okamoto Y, Casazza JP, Kuruppu J, Kunstman K, Wolinsky S, Grossman Z, Dybul M, Oxenius A, Price DA, Connors M, Koup RA. 2002. HIV preferentially infects HIV-specific CD4+ T cells. Nature 417:95–98. doi: 10.1038/417095a [DOI] [PubMed] [Google Scholar]
  • 315. Furch BD, Koethe JR, Kayamba V, Heimburger DC, Kelly P. 2020. Interactions of Schistosoma mansoni and HIV in sub-Saharan Africa: a systematic review. Am J Trop Med Hyg 102:711–718. doi: 10.4269/ajtmh.19-0494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316. Secor WE. 2012. The effects of schistosomiasis on HIV/AIDS infection, progression and transmission. Curr Opin HIV AIDS 7:254–259. doi: 10.1097/COH.0b013e328351b9e3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317. Downs JA, Dupnik KM, van Dam GJ, Urassa M, Lutonja P, Kornelis D, de Dood CJ, Hoekstra P, Kanjala C, Isingo R, Peck RN, Lee MH, Corstjens P, Todd J, Changalucha JM, Johnson WD, Fitzgerald DW. 2017. Schistosoma mansoni and HIV infection in a Ugandan population with high HIV and helminth prevalence. PLoS Negl Trop Dis 11:e0005968. doi: 10.1371/journal.pntd.0005968 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318. Ciccone EJ, Greenwald JH, Lee PI, Biancotto A, Read SW, Yao MA, Hodge JN, Thompson WL, Kovacs SB, Chairez CL, Migueles SA, Kovacs JA, Margolis LB, Sereti I. 2011. CD4+ T cells, including Th17 and cycling subsets, are intact in the gut mucosa of HIV-1-infected long-term nonprogressors. J Virol 85:5880–5888. doi: 10.1128/JVI.02643-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319. Midzi N, Mduluza T, Mudenge B, Foldager L, Leutscher PDC. 2017. Decrease in seminal HIV-1 RNA load after praziquantel treatment of urogenital cchistosomiasis coinfection in HIV-positive men—an observational study. Open Forum Infect Dis 4:ofx199. doi: 10.1093/ofid/ofx199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 320. Yegorov S, Joag V, Galiwango RM, Good SV, Mpendo J, Tannich E, Boggild AK, Kiwanuka N, Bagaya BS, Kaul R. 2019. Schistosoma mansoni treatment reduces HIV entry into cervical CD4+ T cells and induces IFN-I pathways. Nat Commun 10:2296. doi: 10.1038/s41467-019-09900-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 321. Chen Y, Lin H, Cole M, Morris A, Martinson J, Mckay H, Mimiaga M, Margolick J, Fitch A, Methe B, Srinivas VR, Peddada S, Rinaldo CR. 2021. Signature changes in gut microbiome are associated with increased susceptibility to HIV-1 infection in MSM. Microbiome 9:237. doi: 10.1186/s40168-021-01168-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 322. Woodall PA, Kramer MR. 2018. Schistosomiasis and Infertility in East Africa. Am J Trop Med Hyg 98:1137–1144. doi: 10.4269/ajtmh.17-0280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 323. Plesniarski A, Siddik AB, Su RC. 2021. The microbiome as a key regulator of female genital tract barrier function. Front Cell Infect Microbiol 11:790627. doi: 10.3389/fcimb.2021.790627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324. Brenchley JM, Douek DC. 2008. HIV infection and the gastrointestinal immune system. Mucosal Immunol 1:23–30. doi: 10.1038/mi.2007.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 325. Dzhivhuho GA, Rehrl SA, Ndlovu H, Horsnell WGC, Brombacher F, Williamson A-L, Chege GK, Silvestri G. 2018. Chronic schistosomiasis suppresses HIV-specific responses to DNA-MVA and MVA-gp140 Env vaccine regimens despite antihelminthic treatment and increases helminth-associated pathology in a mouse model. PLoS Pathog 14:e1007182. doi: 10.1371/journal.ppat.1007182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326. Mouser EEIM, Pollakis G, Paxton WA. 2012. Effects of helminths and Mycobacterium tuberculosis infection on HIV-1: a cellular immunological perspective. Curr Opin HIV AIDS 7:260–267. doi: 10.1097/COH.0b013e3283521144 [DOI] [PubMed] [Google Scholar]
  • 327. Gazzinelli-Guimaraes PH, Nutman TB. 2018. Helminth parasites and immune regulation. F1000Res 7:1685. doi: 10.12688/f1000research.15596.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328. Kallestrup P, Zinyama R, Gomo E, Butterworth AE, van Dam GJ, Erikstrup C, Ullum H. 2005. Schistosomiasis and HIV-1 infection in rural Zimbabwe: implications of coinfection for excretion of eggs. J INFECT DIS 191:1311–1320. doi: 10.1086/428907 [DOI] [PubMed] [Google Scholar]
  • 329. Mbabazi PS, Andan O, Fitzgerald DW, Chitsulo L, Engels D, Downs JA, Carvalho EM. 2011. Examining the relationship between urogenital schistosomiasis and HIV infection. PLoS Negl Trop Dis 5:e1396. doi: 10.1371/journal.pntd.0001396 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 330. Colombe S, Corstjens P, de Dood CJ, Miyaye D, Magawa RG, Mngara J, Kalluvya SE, van Lieshout L, van Dam GJ, Downs JA. 2018. HIV-1 viral loads are not elevated in individuals co-infected with Schistosoma mansoni spp. After adjustment for duration of HIV-1 infection. Front Immunol 9:2005. doi: 10.3389/fimmu.2018.02005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331. Colombe S, Machemba R, Mtenga B, Lutonja P, Kalluvya SE, de Dood CJ, Hoekstra PT, van Dam GJ, Corstjens P, Urassa M, Changalucha JM, Todd J, Downs JA. 2018. Impact of schistosome infection on long-term HIV/AIDS outcomes. PLoS Negl Trop Dis 12:e0006613. doi: 10.1371/journal.pntd.0006613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332. Yang Y, Xiao P-L, Yang Y, Gao J-C, Shi Y, Cheng W-T, Chen Y, Song X-X, Jiang Q-W, Zhou Y-B. 2018. Immune dysfunction and coinfection with human immunodeficiency virus and Schistosoma japonicum in Yi people. J Immunol Res 2018:6989717. doi: 10.1155/2018/6989717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333. Mazigo HD, Dunne DW, Morona D, Lutufyo TE, Kinung’hi SM, Kaatano G, Nuwaha F. 2015. Periportal fibrosis, liver and spleen sizes among S. mansoni mono or co-infected individuals with human immunodeficiency virus-1 in fishing villages along Lake Victoria shores, North-Western, Tanzania. Parasit Vectors 8:260. doi: 10.1186/s13071-015-0876-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334. Ocama P, Opio KC, Seremba E, Ajal P, Apica BS, Aginya EO. 2017. The burden, pattern and factors that contribute to periportal fibrosis in HIV-infected patients in an S. mansoni endemic rural Uganda. Afr Health Sci 17:301–307. doi: 10.4314/ahs.v17i2.2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 335. Marti AI, Colombe S, Masikini PJ, Kalluvya SE, Smart LR, Wajanga BM, Jaka H, Peck RN, Downs JA, Friedman JF. 2017. Increased hepatotoxicity among HIV-infected adults co-infected with Schistosoma mansoni in Tanzania: a cross-sectional study. PLoS Negl Trop Dis 11:e0005867. doi: 10.1371/journal.pntd.0005867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336. Colombe S, Lee MH, Masikini PJ, van Lieshout L, de Dood CJ, Hoekstra PT, Corstjens PLAM, Mngara J, van Dam GJ, Downs JA. 2018. Decreased sensitivity of Schistosoma mansoni sp. egg microscopy in women and HIV-infected individuals. Am J Trop Med Hyg 98:1159–1164. doi: 10.4269/ajtmh.17-0790 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337. Sanya RE, Muhangi L, Nampijja M, Nannozi V, Nakawungu PK, Abayo E, Webb EL, Elliott AM, LaVIISWA study team . 2015. Schistosoma mansoni and HIV infection in a Ugandan population with high HIV and helminth prevalence. Trop Med Int Health 20:1201–1208. doi: 10.1111/tmi.12545 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338. UNAIDS . 2020. Update: 90–90–90: Good Progress, but the World Is off-Track for Hitting the 2020 Targets. Available from: https://www.unaids.org/en/resources/presscentre/featurestories/2020/september/20200921_90-90-90
  • 339. What is Viral Hepatitis? | CDC. https://www.cdc.gov/hepatitis/abc/index.htm.
  • 340. Jacobsen KH. 2018. Globalization and the changing epidemiology of hepatitis A virus. Cold Spring Harb Perspect Med 8:a031716. doi: 10.1101/cshperspect.a031716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341. Rein DB, Stevens GA, Theaker J, Wittenborn JS, Wiersma ST. 2012. The global burden of hepatitis E virus genotypes 1 and 2 in 2005. Hepatology 55:988–997. doi: 10.1002/hep.25505 [DOI] [PubMed] [Google Scholar]
  • 342. MacLachlan JH, Cowie BC. 2015. Hepatitis B virus epidemiology. Cold Spring Harb Perspect Med 5:a021410. doi: 10.1101/cshperspect.a021410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343. Spearman CW, Dusheiko GM, Hellard M, Sonderup M. 2019. Hepatitis C. The Lancet 394:1451–1466. doi: 10.1016/S0140-6736(19)32320-7 [DOI] [PubMed] [Google Scholar]
  • 344. Sagnelli C, Sagnelli E, Russo A, Pisaturo M, Occhiello L, Coppola N. 2021. HBV/HDV co-infection: epidemiological and clinical changes, recent knowledge and future challenges. Life (Basel) 11:1–17. doi: 10.3390/life11020169 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 345. Nucleoside Analogues . 2012. In Livertox: Clinical and research information on drug-induced liver injury. In National Institute of diabetes and digestive and kidney diseases [PubMed] [Google Scholar]
  • 346. Treem WR, Palmer M, Lonjon-Domanec I, Seekins D, Dimick-Santos L, Avigan MI, Marcinak JF, Dash A, Regev A, Maller E, Patwardhan M, Lewis JH, Rockey DC, Di Bisceglie AM, Freston JW, Andrade RJ, Chalasani N. 2021. Consensus guidelines: best practices for detection, assessment and management of suspected acute drug-induced liver injury during clinical trials in adults with chronic viral hepatitis and adults with cirrhosis secondary to hepatitis B. Drug Saf 44:133–165. doi: 10.1007/s40264-020-01014-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 347. Baumert TF, Berg T, Lim JK, Nelson DR. 2019. Status of direct-acting antiviral therapy for HCV infection and remaining challenges. Gastroenterology 156:431–445. doi: 10.1053/j.gastro.2018.10.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348. Stapleton JT. 1995. Host immune response to hepatitis A virus. J Infect Dis 171 Suppl 1:S9–14. doi: 10.1093/infdis/171.supplement_1.s9 [DOI] [PubMed] [Google Scholar]
  • 349. Shin EC, Sung PS, Park SH. 2016. Immune responses and Immunopathology in acute and chronic viral hepatitis. Nat Rev Immunol 16:509–523. doi: 10.1038/nri.2016.69 [DOI] [PubMed] [Google Scholar]
  • 350. Chatzileontiadou DSM, Sloane H, Nguyen AT, Gras S, Grant EJ. 2020. The many faces of CD4+ T cells: immunological and structural characteristics. Int J Mol Sci 22:1–27. doi: 10.3390/ijms22010073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351. Reina-Campos M, Scharping NE, Goldrath AW. 2021. CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol 21:718–738. doi: 10.1038/s41577-021-00537-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352. Lam JH, Smith FL, Baumgarth N. 2020. B cell activation and response regulation during viral infections. Viral Immunol 33:294–306. doi: 10.1089/vim.2019.0207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353. Xu C, Chen J, Chen X. 2021. Host innate immunity against hepatitis viruses and viral immune evasion. Front Microbiol 12:740464. doi: 10.3389/fmicb.2021.740464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354. Omar HH. 2019. Impact of chronic schistosomiasis and HBV/HCV co-infection on the liver: current perspectives. Hepat Med 11:131–136. doi: 10.2147/HMER.S155962 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355. Mohd Hanafiah K, Groeger J, Flaxman AD, Wiersma ST. 2013. Global epidemiology of hepatitis C virus infection: new estimates of age-specific antibody to HCV seroprevalence. Hepatology 57:1333–1342. doi: 10.1002/hep.26141 [DOI] [PubMed] [Google Scholar]
  • 356. Omar HH, Taha SA, Hassan WH, Omar HH. 2017. Impact of schistosomiasis on increase incidence of occult hepatitis B in chronic hepatitis C patients in Egypt. J Infect Public Health 10:761–765. doi: 10.1016/j.jiph.2016.11.010 [DOI] [PubMed] [Google Scholar]
  • 357. Khatami A, Bahadory S, Ghorbani S, Saadati H, Zarei M, Soleimani A, Zibaei M. 2021. Two rivals or colleagues in the liver? Hepatit B virus and Schistosoma mansoni co-infections: a systematic review and meta-analysis. Microb Pathog 154:104828. doi: 10.1016/j.micpath.2021.104828 [DOI] [PubMed] [Google Scholar]
  • 358. Zhang Y, Xie Y, Chen Q, Chen X, Dong Z, Tan X. 2020. Prevalence and co-infection of schistosomiasis/hepatitis B among rural populations in endemic areas in Hubei, China. Trans R Soc Trop Med Hyg 114:155–161. doi: 10.1093/trstmh/trz086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359. Mavilia MG, Wu GY. 2018. HBV-HCV coinfection: viral interactions, management, and viral reactivation. J Clin Transl Hepatol 6:296–305. doi: 10.14218/JCTH.2018.00016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360. El-Shabrawi MH, Kamal NM. 2013. Burden of pediatric hepatitis C. World J Gastroenterol 19:7880–7888. doi: 10.3748/wjg.v19.i44.7880 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 361. Abruzzi A, Fried B, Alikhan SB. 2016. Coinfection of Schistosoma mansoni species with hepatitis B or hepatitis C viruses. Adv Parasitol 91:111–231. doi: 10.1016/bs.apar.2015.12.003 [DOI] [PubMed] [Google Scholar]
  • 362. Abdel-Rahman M, El-Sayed M, El Raziky M, Elsharkawy A, El-Akel W, Ghoneim H, Khattab H, Esmat G. 2013. Coinfection with hepatitis C virus and schistosomiasis: fibrosis and treatment response. World J Gastroenterol 19:2691–2696. doi: 10.3748/wjg.v19.i17.2691 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363. Van-Lume DS de M, Albuquerque M de FPM de, Souza AI de, Domingues ALC, Lopes EP de A, Morais CNL de, Montenegro SML. 2013. Association between Schistosomiasis mansoni and hepatitis C: systematic review. Rev Saude Publica 47:414–424. doi: 10.1590/S0034-910.2013047004247 [DOI] [PubMed] [Google Scholar]
  • 364. Gasim GI, Bella A, Adam I. 2015. Schistosomiasis, hepatitis B and hepatitis C co-infection. Virol J 12:19. doi: 10.1186/s12985-015-0251-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365. Muir R, Metcalf T, Fourati S, Bartsch Y, Kyosiimire-Lugemwa J, Canderan G, Alter G, Muyanja E, Okech B, Namatovu T, et al. 2023. Schistosoma mansoni infection alters the host pre-vaccination environment resulting in blunted hepatitis B vaccination immune responses. PLoS Negl Trop Dis 17:e0011089. doi: 10.1371/journal.pntd.0011089 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366. Bullington BW, Klemperer K, Mages K, Chalem A, Mazigo HD, Changalucha J, Kapiga S, Wright PF, Yazdanbakhsh MM, Downs JA. 2021. Effects of schistosomes on host anti-viral immune response and the acquisition, virulence, and prevention of viral infections: a systematic review. PLoS Pathog 17:e1009555. doi: 10.1371/journal.ppat.1009555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J, Finn RS. 2021. Hepatocellular carcinoma. Nat Rev Dis Primers 7:1–28. doi: 10.1038/s41572-020-00240-3 [DOI] [PubMed] [Google Scholar]
  • 368. Gohary AA, Elyamany A, Abdalla D, Abolfadl A. 2020. Comparison of hepatic and splenic stiffness in chronic hepatitis C infection with and without schistosomal infection; correlation with hepatic histopathological changes. Clin Exp Hepatol 6:275–281. doi: 10.5114/ceh.2020.99529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 369. Loffredo-Verde E, Bhattacharjee S, Malo A, Festag J, Kosinska AD, Ringelhan M, Rim Sarkar S, Steiger K, Heikenwaelder M, Protzer U, Prazeres da Costa CU. 2020. Dynamic, helminth-induced immune modulation influences the outcome of acute and chronic hepatitis B virus infection. J Infect Dis 221:1448–1461. doi: 10.1093/infdis/jiz594 [DOI] [PubMed] [Google Scholar]
  • 370. Marchese V, Beltrame A, Angheben A, Marocco S, Gaeta GB, Bisoffi Z. 2020. The impact of schistosomiasis co-infection in the presentation of viral hepatitis B in migrants: an observational study in non-endemic area. Travel Med Infect Dis 35:101467. doi: 10.1016/j.tmaid.2019.101467 [DOI] [PubMed] [Google Scholar]
  • 371. Rizzo GEM, Cabibbo G, Craxì A. 2022. Hepatitis B virus-associated hepatocellular carcinoma. Viruses 14:986. doi: 10.3390/v14050986 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 372. Andrade JR, Silva LD, Guimaraes CM, Bassetti-Soares E, Cambraia RD, Couto OFM, Teixeira R. 2014. Chronic hepatitis B and liver Schistosomiasis: A deleterious Association. Transactions of the Royal Society of Tropical Medicine and Hygiene 108:159–164. doi: 10.1093/trstmh/tru010 [DOI] [PubMed] [Google Scholar]
  • 373. Badr RM, Mahomed FF, Fattah SA, Strickland GT, Kamel M, Ghaffar YA. 1991. The impact of endemic schistosomiasis on acute viral hepatitis. Am J Trop Med Hyg 45:743–750. doi: 10.4269/ajtmh.1991.45.743 [DOI] [PubMed] [Google Scholar]
  • 374. Azer SA, Limaiem F. 2023. Cytomegalovirus colitis [PubMed]
  • 375. Zuhair M, Smit GSA, Wallis G, Jabbar F, Smith C, Devleesschauwer B, Griffiths P. 2019. Estimation of the worldwide seroprevalence of cytomegalovirus: a systematic review and meta-analysis. Rev Med Virol 29:e2034. doi: 10.1002/rmv.2034 [DOI] [PubMed] [Google Scholar]
  • 376. Cannon MJ, Schmid DS, Hyde TB. 2010. Review of cytomegalovirus seroprevalence and demographic characteristics associated with infection. Rev Med Virol 20:202–213. doi: 10.1002/rmv.655 [DOI] [PubMed] [Google Scholar]
  • 377. About Cytomegalovirus and Congenital CMV Infection | CDC. https://www.cdc.gov/cmv/overview.html.
  • 378. van Zuylen WJ, Hamilton ST, Naing Z, Hall B, Shand A, Rawlinson WD. 2014. Congenital cytomegalovirus infection: clinical presentation, epidemiology, diagnosis and prevention. Obstet Med 7:140–146. doi: 10.1177/1753495X14552719 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 379. Congenital CMV and Hearing Loss | CDC. https://www.cdc.gov/cmv/hearing-loss.html.
  • 380. Lérias JR, Paraschoudi G, Silva I, Martins J, de Sousa E, Condeço C, Figueiredo N, Carvalho C, Dodoo E, Jäger E, Rao M, Maeurer M. 2019. Clinically relevant immune responses against cytomegalovirus: implications for precision medicine. IJMS 20:1986. doi: 10.3390/ijms20081986 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 381. Cicin-Sain L. 2019. Cytomegalovirus memory inflation and immune protection. Med Microbiol Immunol 208:339–347. doi: 10.1007/s00430-019-00607-8 [DOI] [PubMed] [Google Scholar]
  • 382. Forte E, Zhang Z, Thorp EB, Hummel M. 2020. Cytomegalovirus latency and reactivation: an intricate interplay with the host immune response. Front Cell Infect Microbiol 10:130. doi: 10.3389/fcimb.2020.00130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 383. Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. 2002. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science 296:1323–1326. doi: 10.1126/science.1070884 [DOI] [PubMed] [Google Scholar]
  • 384. Gao Y, Zhang X, Jiang T, Zhou H, Liu H, Hu Y, Cao J, Hsieh MH. 2023. Inhibition of hepatic natural killer cell function via the TIGIT receptor in schistosomiasis-induced liver fibrosis. PLoS Pathog 19:e1011242. doi: 10.1371/journal.ppat.1011242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 385. Hou X, Yu F, Man S, Huang D, Zhang Y, Liu M, Ren C, Shen J. 2012. Negative regulation of Schistosoma japonicum egg-induced liver fibrosis by natural killer cells. PLoS Negl Trop Dis 6:e1456. doi: 10.1371/journal.pntd.0001456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 386. Arbovirus - an overview | ScienceDirect Topics. https://www.sciencedirect.com/topics/medicine-and-dentistry/arbovirus.
  • 387. Madewell ZJ. 2020. Arboviruses and their vectors. South Med J 113:520–523. doi: 10.14423/SMJ.0000000000001152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 388. Viglietta M, Bellone R, Blisnick AA, Failloux AB. 2021. Vector specificity of arbovirus transmission. Front Microbiol 12:773211. doi: 10.3389/fmicb.2021.773211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 389. Rodhain F. 1998. The idea of natural reservoir in arbovirology. Bull Soc Pathol Exot 91:279–282. [PubMed] [Google Scholar]
  • 390. Atkinson B, Hewson R. 2018. Emerging arboviruses of clinical importance in Central Asia. J Gen Virol 99:1172–1184. doi: 10.1099/jgv.0.001125 [DOI] [PubMed] [Google Scholar]
  • 391. Rocklöv J, Dubrow R. 2020. Author correction: climate change: an enduring challenge for vector-borne disease prevention and control control (Nature Immunology, (2020), 21, 5, (479-483), 10.1038/s41590-020-0648-y). Nat Immunol 21:479–483. doi: 10.1038/s41590-020-0692-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 392. Campbell-Lendrum D, Manga L, Bagayoko M, Sommerfeld J. 2015. Climate change and vector-borne diseases: what are the implications for public health research and policy?. Philos Trans R Soc Lond B Biol Sci 370:20130552. doi: 10.1098/rstb.2013.0552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393. Higuera A, Ramírez JD. 2019. Molecular epidemiology of dengue, yellow fever, Zika and Chikungunya arboviruses: an update. Acta Trop 190:99–111. doi: 10.1016/j.actatropica.2018.11.010 [DOI] [PubMed] [Google Scholar]
  • 394. CDC - Schistosomiasis - FAQs. https://www.cdc.gov/parasites/schistosomiasis/gen_info/faqs.html.
  • 395. Ogunlade ST, Meehan MT, Adekunle AI, Rojas DP, Adegboye OA, McBryde ES. 2021. A review: Aedes-borne arboviral infections, controls and Wolbachia-based strategies. Vaccines 9:32. doi: 10.3390/vaccines9010032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 396. Overgaard HJ, Dada N, Lenhart A, Stenström TAB, Alexander N. 2021. Integrated disease management: arboviral infections and Waterborne diarrhoea. Bull World Health Organ 99:583–592. doi: 10.2471/BLT.20.269985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397. Deol AK, Fleming FM, Calvo-Urbano B, Walker M, Bucumi V, Gnandou I, Tukahebwa EM, Jemu S, Mwingira UJ, Alkohlani A, Traoré M, Ruberanziza E, Touré S, Basáñez M-G, French MD, Webster JP. 2019. Schistosomiasis — assessing progress toward the 2020 and 2025 global goals. N Engl J Med 381:2519–2528. doi: 10.1056/NEJMoa1812165 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 398. Alves LS, Estanislau C, Barreto L, Batista F, Toppa N. 2017. Concomitant testicular infection by Zika virus and Schistosoma mansoni in a Brazilian young boy. Rev Assoc Med Bras 63:500–503. doi: 10.1590/1806-9282.63.06.500 [DOI] [PubMed] [Google Scholar]
  • 399. Ahmed AFM, El-Sayad MH, Ali HS, El-Taweel HA. 2021. Impact of coinfection with Schistosoma mansoni on the antibody response to Helicobacter pylori. Acta Parasit 66:857–862. doi: 10.1007/s11686-020-00330-y [DOI] [PubMed] [Google Scholar]
  • 400. Rosadas C, Taylor GP. 2022. HTLV-1 and Co-infections. Front. Med 9. doi: 10.3389/fmed.2022.812016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 401. Pielok Ł, Frąckowiak K, Kłudkowska M. 2019. Disseminated skin rash and blood eosinophilia in a Polish traveler diagnosed Strongyloides stercoralis, Trichuris trichiura, Schistosoma mansoni sp. and Blastocystis sp. coinfection. Ann Parasitol 65:99–102. doi: 10.17420/ap6501.188 [DOI] [PubMed] [Google Scholar]
  • 402. Fiamma M, Longoni SS, Siddig EE, Attene S, Astone V, Nicoletti A, Paglietti B, Santona A, Fele A, Ivaldi R, Logias F. 2020. Multi-parasite infection in an immigrant from Ghana: potential for new epidemic foci. J Infect Dev Ctries 14:1344–1348. doi: 10.3855/jidc.13151 [DOI] [PubMed] [Google Scholar]
  • 403. McLaughlin TA, Nizam A, Hayara FO, Ouma GS, Campbell A, Khayumbi J, Ongalo J, Ouma SG, Shah NS, Altman JD, Kaushal D, Rengarajan J, Ernst JD, Blumberg HM, Waller LA, Gandhi NR, Day CL, Benkeser D. 2021. Schistosoma mansoni infection is associated with a higher probability of tuberculosis disease in HIV-infected adults in Kenya. J Acquir Immune Defic Syndr 86:157–163. doi: 10.1097/QAI.0000000000002536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 404. Gordon MA, Zijlstra EE, Naus CWA, Visser LG, Molyneux ME, van Lieshout L. 2003. Schistosomiasis does not contribute to death or recurrence of nontyphoid Salmonella bacteremia in human immunodeficiency virus-infected Malawian adults. Clin Infect Dis 37:e177–e179. doi: 10.1086/379828 [DOI] [PubMed] [Google Scholar]
  • 405. Dunne DW, Cooke A. 2005. A worm’s eye view of the immune system: consequences for evolution of human autoimmune disease. Nat Rev Immunol 5:420–426. doi: 10.1038/nri1601 [DOI] [PubMed] [Google Scholar]
  • 406. Osakunor DNM, Ishida K, Lamanna OK, Rossi M, Dwomoh L, Hsieh MH, Cwiklinski K. 2022. Host tissue proteomics reveal insights into the molecular basis of Schistosoma haematobium-induced bladder pathology. PLoS Negl Trop Dis 16:e0010176. doi: 10.1371/journal.pntd.0010176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 407. Buck JC, De Leo GA, Sokolow SH. 2020. Concomitant immunity and worm senescence may drive schistosomiasis epidemiological patterns: an ECO-evolutionary perspective. Immunol 11:490788. doi: 10.3389/fimmu.2020.00160 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical Microbiology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES