Immune Response To Schistosoma

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Parasite Immunology, 2014, 36, 428–438 DOI: 10.1111/pim.

12084

Review Article

Immune responses to Schistosoma haematobium infection

J. I. ODEGAARD1 & M. H. HSIEH2

1
Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA, 2Department of Urology, Stanford University
School of Medicine, Stanford, CA, USA

SUMMARY tentive funding agencies and the difficulties inherent in


studying fastidious organisms with complicated lifecycles.
Urogenital schistosomiasis is one of the greatest single infec-
Nevertheless, recently refocused philanthropic and govern-
tious sources of human morbidity and mortality known.
mental funding has enabled several critical advancements
Through a complex cycle of infection, migration and even-
in our ability to experimentally manipulate S. haematobi-
tual maturation and mating, S. haematobium (the aetiologi-
um, opening the species-specific biology of this important
cal agent of urogenital schistosomiasis) deposits highly
organism to systematic study for the first time.
immunogenic eggs within the bladder and other pelvic
In this review, we discuss the recent evolution of S. hae-
organs, activating a wide range of immune programs that
matobium-specific reagents and techniques and how these
determine both infection outcome as well as downstream
complement traditional clinical studies to describe the
immunopathology. In this review, we discuss the experimen-
unique pathology of S. haematobium infection. More spe-
tal and observational bases for our current understanding of
cifically, we will focus on the complex immune response to
these immune programs, focusing specifically on how the
urogenital egg deposition and discuss how the parasite-
balance of type 1 and type 2 responses governs subsequent
host interaction evolves to either parasite elimination or
immunopathology and clinical outcome.
progressive clinical disease.
Keywords immune modulation, immunoepidemiology, Schis-
tosoma haematobium, Schistosoma spp, schistosomiasis, SCHISTOSOMA HAEMATOBIUM BIOLOGY
urogenital
Like other schistosomes, S. haematobium occupies a com-
plex and specific biological niche (reviewed in (4)). The
INTRODUCTION lifecycle begins in fresh water aquatic environments, where
eggs hatch into miracidia and infest various species of
With well over one-half billion people affected worldwide,
freshwater snail (Bulinus), which act as the intermediate
schistosomiasis is one of the most wide-reaching clinically
host. Upon maturation, S. haematobium emerges as free-
significant infections on the planet (1,2). Moreover, Schis-
swimming cercariae that traverse the water column until
tosoma haematobium, as the most prevalent member of the
they come into contact with human skin, into which they
genus, is one of the greatest single sources of human
burrow. With access to the dermal vasculature, the cerca-
morbidity and mortality known with an African disease
riae mature to schistosomulae, migrate through the host
burden that equals and may possibly exceed even that of
circulation, mature and eventually lodge in specific venous
malaria (3). Despite such impressive infamy, much of
plexuses within the body as adult worm mating pairs and
S. haematobium’s biology and pathology remains
begin to lay eggs. Importantly, this migration is not at all
surprisingly enigmatic, due in perhaps equal parts to silent
random and instead demonstrates remarkable tissue speci-
infections, a decades-long clinical course, historically inat-
ficity; S. haematobium, for example, migrates to venous
plexuses in the pelvis (primarily those draining the bladder
Correspondence: Michael H. Hsieh, Department of Urology, Stan- and female genital tract), whereas S. mansoni and
ford University School of Medicine, Stanford, CA 94305, USA S. japonicum lodge in the portal circulation. This anatomi-
(e-mail: mhhsieh@stanford.edu).
Disclosures: None.
cal particularity is crucial to understanding the varied clin-
Received: 24 May 2013 ical manifestations associated with schistosome infection
Accepted for publication: 20 October 2013 as it tethers subsequent pathology to specific tissues,

428 © 2013 John Wiley & Sons Ltd


Volume 36, Number 9, September 2014 Immune responses to S. haematobium infection

setting the stage for hepatosplenic (as in S. mansoni and routine use. Due to this experimental intransigence, much
S. japonicum infection) or urogenital disease (as in S. hae- of schistosome-directed research has focused instead on
matobium infection). Once in place, adult worm pairs may rodent models of S. mansoni and, to a lesser extent, S. ja-
persist for many fecund years and produce thousands of ponicum, both of which approximate human hepatosplenic
eggs. Though many eggs eventually pass into the intestinal disease in the mouse. As these models have established
or bladder lumens and are excreted, many become trapped important paradigms that apply to S. haematobium as
within the host tissue, evoking a potent immune response well, mounting evidence suggests that species- and tissue-
that, over months and years, results in dramatic tissue specific factors make such approaches poor surrogates in
remodelling and long-term pathological consequences. the study of urogenital schistosomiasis. For example, the
Much of our knowledge of the pathophysiology of effects of egg expulsion are very different in the bacteria-
schistosome infections is derived from two basic rich environment of the gut lumen when compared with
approaches: clinical observation and animal models. Clini- the sterile bladder, just as the biochemical functionality of
cal observation has formed the basis for urogenital schis- the liver is affected differently than the biomechanical
tosomiasis research since its inception, generally in the function of the bladder. Indeed, even within the same
form of serological/urinary analyses (antibody responses pathological context of hepatosplenic disease, S. haemato-
or hematuria, for example), ultrasound examination, or in bium, S. mansoni and S. japonicum infections all result in
monitoring end pathology (such as renal failure or urothe- different histological appearances, pathological sequelae
lial carcinoma), all of which lend valuable insights into the and disease kinetics.
natural history of chronic infection including the various As S. haematobium fails to establish urogenital disease
possible immunopathological outcomes. Moreover, studies in all but a small fraction of infected mice (5,6), studies of
involving urine and serum analysis provide a serial view of mouse hepatic schistosomiasis demonstrate that the basic
infection evolution over time. Neither of these approaches, host immune response is intact and similar to that of
however, is able to provide the standardization or experi- human hepatosplenic disease, strongly supporting the gen-
mental manipulability necessary for rigorous definition or eral applicability of this model of infection. To circumvent
mechanistic investigation. Moreover, the absence of acute S. haematobium’s lack of pelvic organ involvement in the
clinical stigmata of infection largely precludes identifica- mouse, our laboratory developed a novel model of urogen-
tion of recently infected individuals and, thus, study of the ital schistosomiasis in which S. haematobium eggs—the
nascent phases of infection including those early determi- primary determinants of the host immune responses—are
nants of resistance to chronic infection. surgically implanted into the mouse bladder lamina pro-
Where heterogeneous patient backgrounds and environ- pria (the layer of loose connective tissue between the uro-
ments, unknown infection duration and ethical restrictions thelium and the smooth muscle in which eggs lodge in
on intervention complicate human studies, animal models human infection), effectively simulating bladder oviposi-
are relatively free of these shortcomings as their genetic tion in urogenital disease (7). Importantly, this model
backgrounds, environmental exposures and infection tim- faithfully recapitulates the salient architectural, pathologi-
ing may be rigorously controlled and manipulated. cal and immunological sequelae of human disease, thus
Because of this tractability, animal models are indispens- offering the first experimentally tractable model of urogen-
able in infectious agent research; however, care must be ital schistosomiasis. This model in combination with
taken to ensure faithful recapitulation of human disease. recent in vitro technological advances (such as S. haemato-
Importantly, animal model fidelity has proven to be par- bium-directed short interfering RNA technology (8)) and
ticularly problematic in the study of urogenital schistoso- bioinformatic resources (such as the publication of the
miasis. For most disease research, mice are the favoured S. haematobium genome (9)) has, for the first time, placed
model organism—their genetics are both defined and many of the fundamental questions in schistosomal uro-
manipulable, they breed and subsist successfully in con- genital immunity and pathology within reach.
strained lab environments, and a bevy of species-specific
tools and experience is available for them. In an unfortu-
INNATE IMMUNE RESPONSES TO
nate contrast to human disease, however, S. haematobium
SCHISTOSOME INFECTION
worm pairs exhibit a tropism for the portal circulation in
the mouse and other commonly employed rodents, elicit- As discussed above, schistosome infections begin with
ing pathology quite distinct from urogenital disease. cercarial penetration of the skin, migration into the der-
Indeed, researchers have had to resort to costly and ethi- mal vasculature and eventual deposition in a terminal
cally fraught primate models to approximate human uro- venous plexus, the location of which determines the specif-
genital pathology, none of which have proven tenable for ics of the eventual clinical disease. During this weeks-long

© 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438 429
J. I. Odegaard & M. H. Hsieh Parasite Immunology

migration, cercariae mature into adult worms and form (a)


male-female pairs, offering the host immune system a
potentially broad array of antigens. Despite this antigenic
diversity, however, the host immune response to cercarial
and adult worm antigens is, by comparison to later egg-
triggered disease, muted (10–13). Indeed, aside from rare
cases of cercarial dermatitis (swimmer’s itch) and Katay-
ama syndrome (hypersensitivity responses to acute and
mid-stage infection, respectively, both of which are espe-
cially rare in S. haematobium infection relative to other
schistosome species), S. haematobium infection is rarely
clinically apparent until egg deposition occurs (as high-
lighted in (4)). Importantly, the immune response to adult
worms, while less intense than that to tissue-embedded
eggs, is the primary determinant of infection outcome,
with those individuals who mount robust antibody (partic- (b)
ularly IgE) responses to adult worm antigens more likely
to resist reinfection than others with more tepid responses
(14).
In contrast to the relatively ineffectual response to via-
ble cercariae and adult worms, the eggs produced by adult
S. haematobium evoke a robust immune response. Unfor-
tunately, early immune responses to egg deposition are lar-
gely undefined in the human due to our inability to
identify and study newly infected individuals. The advent
of the mouse model of direct oviposition (7), however,
opens this response accessible to study. In this context,
S. haematobium eggs elicit an immediate (within 24 h) and
brisk host response from local and recruited innate
immune cells. Interestingly, the character of the initial Figure 1 Schistosoma haematobium egg granuloma structure.
response is mixed, with induction of inflammatory media- (a) Within 4 days, the immune response to surgically implanted
S. haematobium eggs (arrows) forms a loosely organized mass
tors (such as TNF-a) as well as cytokines associated with
lesion (dashed line) within the bladder lamina propria dominated
type 2 responses (such as CCL11). As with many other by dyshesive macrophages admixed with eosinophils, lymphocytes
infectious stimuli, such immediate responses have two sali- and neutrophils. (b) Over time, this response evolves into a highly
ent effects—additional innate immune cells are recruited organized granuloma (dashed line), which comprises a core of
to maintain and amplify the initial response, whereas the multinucleated syncytial macrophages (giant cells) and tightly
cohesive single macrophages surrounding the now-calcified eggs
adaptive immune system is recruited and activated.
(arrows) with admixed eosinophils and peripheral lymphoid
Innate immune cell recruitment is, of course, a hallmark aggregates (asterisks). These features are histologically identical to
of many immune responses; however, schistosome egg-elic- those observed in human infection.
ited responses are distinguished by their degree and persis-
tence—indeed, the recruitment of innate immune cells to
sites of schistosome egg deposition results in the develop- Characteristically, eggs are engulfed by multinucleated syn-
ment of a highly specialized secondary immune tissue cytial macrophages (giant cells), which are in turn sur-
known as a granuloma (Figure 1), an especially impressive rounded by an organized and cohesive cuff of epithelioid
feat in the bladder given the relative paucity of leucocytes macrophages with admixed eosinophils and, to varying
in uninfected tissue. At time points as early as 4 days after degrees, neutrophils concentrated around the granuloma
oviposition in the mouse model (7), S. haematobium eggs periphery. Within the mouse urogenital model, this archi-
are segregated from surrounding tissue by a dense but tecture persists for more than 100 days after oviposition
loosely cohesive aggregate of newly recruited macrophages with minimal but progressive loss of cellularity similar to
and eosinophils. Over the subsequent 7 days, this highly what is observed in mouse models of hepatosplenic dis-
cellular innate immune admixture organizes into a cohe- ease. Most importantly, this response is histologically iden-
sive granuloma centred on the inciting schistosome egg(s). tical to the egg-centred granulomata observed in human

430 © 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438
Volume 36, Number 9, September 2014 Immune responses to S. haematobium infection

disease (15–19), although the polypoid lesions occasionally eosinophils; however, the overall significance of the rat
seen in human disease (20) are not apparent. infection phenotype is unclear and may, in fact, be an idi-
osyncrasy of the model itself.) Upon urogenital oviposi-
tion, however, eosinophils rapidly migrate to the bladder,
Macrophages
presumably in response to CCL5/RANTES), CCL11/
As the primary structural constituent of the granuloma, eotaxin-1 and CCL24/eotaxin-2 (although CCL11 is the
macrophages unsurprisingly play a central role initiating only one of these chemokines known to be highly
and guiding the overall innate immune response. In vitro expressed in the immediate response), where they accumu-
and in vivo experiments have long ago established the mac- late as the most numerous cell type within the granuloma
rophage’s ability to mount robust type 2 responses to (although due to their larger size, macrophages occupy far
schistosome eggs including expression of high levels of more volume) (7). Indeed, in schistosome-infected humans,
ARG1, MRC and CHI3L3 (reviewed in (21)). Indeed, eosinophils are even detectable (albeit at much reduced
S. mansoni infection of mice deficient in type 2 macro- numbers) near old, long-calcified egg remnants in long-
phage responses (such as mice lacking IL-4Ra in macro- standing lesions where much of the granulomatous
phages) results in fatal inflammatory responses due to response has long since disappeared (18,19) (Odegaard,
their inability to form proper granulomata to segregate unpublished observations). Though their functional contri-
eggs from surrounding tissue (22). Moreover, numerous bution in urogenital schistosomiasis has not yet been
observations of hepatic disease models have implicated described, eosinophils serve as an important source of IL-
type 2 macrophages in the initial fibrotic stabilization of 4 prior to the emergence of the Th2 response in many
the granuloma and subsequent associated tissue fibrosis other immunological contexts (such as in adipose tissue
(23–25). Ex vivo interrogation of granuloma-derived leuco- and skeletal muscle) (32,33) and remain one of the pri-
cytes in the mouse model of urogenital disease, however, mary sources of IL-13 throughout the evolution of hepatic
unexpectedly revealed elevated expression of type 1 macro- granulomatous responses (34). Indeed, eosinophil-derived
phage activation markers (such as NOS2 and CD40) con- IL-13 has been suggested to be an important driver of
current with expression of type 2-associated genes (such as fibrosing responses in hepatosplenic schistosomiasis both
ARG1, MRC and CHI3L3) (26), suggesting that egg directly through activation of hepatic stellate cells and
deposition in this context involves either a mixed pheno- indirectly through the promotion of type 2 macrophage
type or a mixed population of type 1 and type 2 macro- activation (34). Congruent with this hypothesis, mice defi-
phages. Moreover, expression analyses of developing cient in IL-5 demonstrate smaller S. mansoni granulomata
granulomata have described extensive expression of both and significantly reduced liver fibrosis (34). More recently,
type 1- and type 2-associated cytokine and chemokine sig- however, the role of the eosinophil has been called into
nalling pathways (26), further suggesting that urogenital question by the absence of any substantial difference
oviposition triggers both macrophage activation programs. between wild type and DdblGATA or TgPHIL mice (both
of which entirely lack eosinophils but demonstrate pre-
served IL-5 production) during infection with S. mansoni
Eosinophils
(35). Though the absence of a phenotype is striking, these
Eosinophil infiltration and eosinophiluria are cardinal his- studies examined hepatic egg-induced pathology only,
tological and clinical signs of parasitic infection (27,28); which does not exclude a role in the elimination of adult
however, much of this cell’s functional significance worms and cercariae.
remains elusive. In the pre-egg stages of the S. haematobi-
um lifecycle, eosinophils are important effector cells capa-
Dendritic cells
ble of killing cercariae and adult worms in a
degranulation- and IgE-dependent manner (29). In fact, As the principal antigen-presenting cell, dendritic cells
the ability of rats to clear schistosome infections without constitute a critical bridge between early innate and later
developing chronic disease may be attributable, in part, to adaptive immune responses to S. haematobium infection;
the ease with which rat eosinophils degranulate in however, the specifics of this role have not yet been
response to IgE-decorated worms relative to eosinophils described. Indeed, even in well-studied models of S. man-
from mice, an animal susceptible to chronic infection (30). soni infection, dendritic cell activation remains somewhat
(This particular observation may also be due, in part, to enigmatic. Dendritic cells (broadly—if inaccurately—
the absence of high-affinity IgE receptor I—a surface defined in many studies by CD11c expression) are
receptor on human eosinophils involved in IgE-mediated necessary for proper Th2 responses to schistosome egg
anti-schistosome activity in vitro (31)—on mouse deposition (36); however, numerous experiments have

© 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438 431
J. I. Odegaard & M. H. Hsieh Parasite Immunology

demonstrated that even highly immunoreactive schisto- true contribution of NK cells even in relatively well-stud-
some antigen preparations (soluble egg antigen, for exam- ied hepatic schistosomiasis is unclear, and their relevance
ple) fail to elicit canonical maturation of dendritic cells or to urogenital disease is entirely unknown.
expression of activating cytokines or costimulatory mole-
cules in vitro (37–39). Despite this apparent apathy, how-
Basophils
ever, these cells demonstrate a surprisingly potent capacity
to induce an activated Th2 phenotype in na€ıve T cells in a Despite the past use of basophil degranulation as a diag-
CD40-, OX40L- and NFjB activation-dependent manner nostic assay for urogenital schistosomiasis (50), the contri-
(39–42). These apparently contradictory findings have bution of basophils to human urogenital schistosomiasis is
generated multiple proposed novel maturation pathways; entirely unknown (as is true in much of basophil biology).
however, in general, little evidence exists to support their The ability of basophils to produce substantial amounts of
in vivo relevance. One such recently proposed mechanism IL-4 in response to S. mansoni infection in mice long sup-
involves omega-1, a T2 RNase abundant in SEA and ported the belief that these cells innately produce IL-4 in
secreted by viable S. mansoni eggs (43). When internalized response to schistosome exposure and initiate the observed
by immature dendritic cells, this particular molecule drives type 2 bias in the schistosome-associated immune
Th2 polarizing capacity through an ill-defined mechanism response. The description of IL-4 inducing principle from
involving RNase-dependent nonspecific degradation of S. mansoni eggs (IPSE)/alpha-1—an aptly-named protein
host mRNA and rRNA pools (43). How nonspecific RNA secreted from viable schistosome eggs that triggers IL-4
degradation is linked to T cell stimulation and Th2 polari- release from basophils—seemed to support this hypothesis
zation capacity is unclear; however, this finding suggests a (51); however, careful dissection of this putative pathway
novel mechanistic link between the observed in vitro and in mice using multiple helminth species including S. man-
in vivo effects of schistosome antigens. Whether such soni recently debunked this theory through three major
mechanisms govern dendritic cell participation in urogeni- observations. First, basophil deletion yielded no defect in
tal disease is entirely unclear (43), as initial studies failed Th2 responses or any other demonstrable infection-related
to detect omega-1 in S. haematobium infection (44). phenotype (51). Second, basophils were incapable of IL-4
production in the absence of T cells (52). Lastly, interac-
tions between basophils and T cells were shown to be
Natural killer cells
kinetically incompatible with antigen presentation and
As the primary and secondary outcomes of schistosome were demonstrated instead to represent a necessary licens-
infections correlate strongly with the relative balance ing process only after which basophils were capable of
between type 1 and type 2 immune responses, there is con- IL-4 production (52). However, basophils were able to res-
siderable interest in how these activation programs are ini- cue the phenotype of mice lacking IL-4 and IL-13 in their
tiated, how their relative contributions are regulated and T cells, suggesting that though it is not required in wild-
in what ways might they be manipulated for therapeutic type mice, basophil-derived IL-4 can functionally replace
effect. As a major source of IFN-c and other type 1-bias- IL-4 production by Th2 cells during S. mansoni infection
ing cytokines, natural killer (NK) cells are of natural inter- (52). As such, the basophil lineage remains a potential
est in this context; however, the data supporting their role contributor to schistosome-associated immune responses
in schistosome infection is limited to three direct observa- and immunopathology; however, the nonredundant func-
tions. First, NK cells are recruited to schistosome egg- tions of this putative role remain unclear.
associated hepatic granulomata (45), where their presence
corresponds to locally increased levels of IL-12 (although
Mast cells
they do not appear to contribute significantly to IFN-c
levels within the granuloma or serum) (46). Second, deple- Due to the strong correlation between serum IgE levels
tion of NK cells results in increased schistosome egg-asso- and resistance to S. haematobium, investigators have long
ciated granuloma volume and fibrosis, whereas their proposed mast cells (one of the primary effector cell types
activation (along with many that of several other lineages) for IgE-dependent parasite killing) as potential mediators
by TLR3 ligands has the opposite effect (46–48). Finally, of IgE’s protective effects. Observational studies in indi-
activated NK cells are capable of killing activated hepatic viduals vocationally exposed to S. mansoni, however, dem-
stellate cells—a key fibrogenic cell in hepatic fibrosis—in onstrated that circulating mast cell precursor numbers
vitro and in vivo in the context of toxin-induced fibrosis were, in fact, negatively correlated with resistance (53),
(49). Though these observations are compelling and sug- although verification of this effect and mechanistic expla-
gest a role in limiting the granulomatous response, the nation are absent. Interestingly, mast cell-derived IL-10

432 © 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438
Volume 36, Number 9, September 2014 Immune responses to S. haematobium infection

was recently implicated in chronic bacterial urinary tract 28 days, whereas Th1- and Th17-associated mediators
infection (54), suggesting a possible role for this lineage in remain equivalent to controls (7). This pattern further
the regulation of chronic inflammatory responses in the holds in mouse models of hepatic disease where the Th1
genitourinary tract and, perhaps, in the eventual induction response acts to limit granuloma size and fibrotic stabiliza-
of regulatory T cell responses. Unsurprisingly, however, lit- tion through both direct and indirect effects (22). Impor-
tle direct data are available regarding the potential role of tantly, serological studies in humans consistently report
mast cells in urogenital schistosomiasis. similar elevations in Th2 cytokines with suppression of
their Th1/Th17 counterparts (63). One important distinc-
tion between the human data and the mouse model of ovi-
ADAPTIVE IMMUNE RESPONSES TO
position, however, resides in the regulatory T cell (Treg)
SCHISTOSOME INFECTION
response. In the mouse model, markers of Treg cells and
responses are suppressed (7), as human reports describe
T cells
increased levels of IL-10 and Treg responses, although at
Though macrophages form the structural bulk of the gran- levels much lower than those of Th2 mediators (63). This
uloma, multiple lines of evidence suggest that T lympho- discrepancy is likely to be due to idiosyncratic rather than
cytes play a critical role in both the local and the systemic species-specific effects as mouse models of S. mansoni
responses to infection. First, T cells rapidly infiltrate egg- infection recapitulate Treg induction, which are required
exposed tissues and are already present in large numbers here for controlling inflammation throughout the process
in the nascent granuloma at day 4 in the mouse model of of egg expulsion into the gut lumen (21). Regardless, there
bladder oviposition (7). In later time points and in human is ample evidence in both mice and humans that schisto-
samples, these cells are found scattered throughout the some soluble egg antigens induce IL-10, which in turn
granuloma as well as concentrated in lymphoid follicles in suppresses T cell responses (64–69). These immunomodu-
the granuloma’s periphery. Second, T cell-deficient mouse latory pathways may ultimately serve to reduce the host
models of S. japonicum and S. mansoni hepatic disease pathophysiology associated with schistosomiasis.
demonstrate egg-centred zones of hepatocyte necrosis Congruent with observations from mouse models, the
accompanied by an exaggerated neutrophil response human data strongly suggest that induction of Th1/Th17
(55,56) suggesting that T cells are required to restrain the mediators, as it is not uniformly correlated with infection,
inflammatory response. Similarly, mouse models of is strongly associated with increased urogenital morbidity
S. mansoni infection require an effective Th2 response to and end-stage pathology, whereas Th2 responses are asso-
sequester highly phlogistic eggs from surrounding tissue ciated with decreased disease sequelae (63). Congruently,
and prevent lethal inflammation (22). Third, both T cells polymorphisms that either impair type 2 (e.g. hypomor-
present locally and in the draining lymph node robustly phic variants in IL13 and STAT6) or enhance type 1
express Th2-associated cytokines (e.g. IL-4 and IL-13) well responses (e.g. hypomorphic variants in CTLA4) are both
in advance of systemic Th2 responses (7), supporting the associated with increased S. haematobium infection inten-
hypothesis that this activation bias is established by early sity in endemic populations (70,71). Taken in aggregate,
T cell responses in the granuloma. Lastly, interference with these data suggest that the balance of type 1 responses rel-
key Th2-restricted factors (such as IL-4) by either genetic ative to type 2 is an important determinant of the local
deletion in mice or polymorphisms in human populations control of urogenital disease and contributes to infection
results in severe restriction of granuloma-forming resistance.
responses and exacerbated disease (22,57–59). These data
in aggregate support a model in which the initial innate
Natural killer T cells
immune response generates Th2 cells, which then support
granuloma formation through IL-4 production, recruit Natural killer T (NKT) cells have not been well studied in
and maintain eosinophils via IL-5, and promote fibrosing the context of schistosome infection; however, studies uti-
responses via IL-13. lizing CD1d / mice, which lack all NKT cells, suggest
In contrast, Th1 and Th17 inflammatory responses are that these cells play a role in type 2/regulatory responses
largely suppressed after an initial spike in the mouse uro- to S. mansoni infections, although direct measures of
genital oviposition model (7). Indeed, S. mansoni infection infection (such as worm and egg counts and hepatic
in mice deficient in IFN-c, IL-12, or IL-17 is little differ- pathology) are unaffected (72). Interestingly, Ja18 / mice,
ent than in wild-type controls (60–62). Congruently, sys- which specifically lack only type I NKT cells, demonstrate
temic levels of type 2 cytokines increase markedly by day impaired type 1 responses whereas type 2/regulatory
7 post-oviposition and remain elevated for at least responses remain intact (72). Moreover, these mice

© 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438 433
J. I. Odegaard & M. H. Hsieh Parasite Immunology

demonstrate larger granulomata, enhanced IgE responses govern resistance to infection, parasite clearance and sus-
and slightly lower egg counts (72), suggesting that NKT ceptibility to re-infection, the proximal determinant of
cell promotion of type 2 responses may be specifically due S. haematobium-related morbidity and mortality is the egg-
to type 2 NKT cells and that type 1 cells may favour type directed immune response. In productive (from S. haemato-
1 responses. This is in contrast to other immunological bium’s point of view) oviposition, the cascade of immune
contexts (such as in adipose tissue and liver metabolism) responses detailed above facilitate the segregation of immu-
and other studies of schistosome infection (73) in which nogenic eggs from the host and shepherd them to the organ
type I NKT cells have been reported to favour type 2/regu- lumen, where they may be expelled from the body and per-
latory responses. As such, the actual contribution of this petuate the helminth’s life cycle. For unclear reasons, how-
lineage to S. mansoni infection is unclear, whereas ever, a subset of eggs fails to be expelled and instead persists
their role in S. haematobium infection remains entirely within the tissue, chronically activating the host immune
undescribed. system. This activation comprises a remarkable diversity of
immune pathways; however, these may be broadly classified
as type 1, type 2, or regulatory responses and are typified by
B cells
their associated T lymphocyte phenotypes with Th1, Th2
Though T lymphocytes play a critical role in managing and Treg activation. Though the human data are far from
schistosome egg-associated pathology, B lymphocytes are clear and are confounded by cohort effects and other biases,
more directly involved in infection clearance and resistance. the eventual outcome of S. haematobium infection is
As in T cells, high levels of S. haematobium-specific type 2 B thought to be directly related to the balance between these
cell effector molecules (primarily IgE) have been associated various immune programs and the tissue responses they
with reduced susceptibility to reinfection (14); however, B direct—Th1 responses are associated with tissue damage
cell-deficient animals are still able to sequester deposited and heightened infection intensity in many human studies,
S. japonicum eggs within granulomata, although with some- whereas Th2 bias is correlated with decreased egg burden
what delayed kinetics (74). In human populations where and infection resistance (56). It should be noted, however,
S. haematobium is endemic, however, B cell responses are that these are general trends and studies exist in each cate-
critical to the development of parasite resistance (14). In gory suggesting contrasting interpretations. The generalized
these populations, exposure is nearly universal as defined by consequences of regulatory responses are similarly unclear
the ubiquity of S. haematobium-specific IgM (75); however, (although control of type 1 responses has been proposed).
chronic disease and end-stage pathology is restricted to a Moreover, it remains unclear whether these responses direct
minority of individuals with a substantial subgroup evincing the disease course or are merely markers thereof. What is
no demonstrable evidence of disease. This population is dis- clear, however, is that as some egg-directed immune
tinguished by several B cell-related criteria including high responses are necessary to protect the host from deleterious
titres of S. haematobium-specific IgE, anti-AWA (adult inflammation and tissue damage during egg transit and
worm antigen) IgG1 and anti-Sh13 IgG3 as well as high expulsion, chronic activation can have marked pathological
IgE:IgG4 (63,76–78). Importantly, these resistance-associ- consequences.
ated immunoglobulin responses are directed not against im- One of the primary sources of morbidity and mortality
munopathogenic egg antigens but against the adult worm. in S. haematobium-infected individuals is egg-induced uri-
It is also important to note, however, that these responses nary tract fibrosis. Here, chronic activation of egg-directed
generally tend to increase with age and that these specific immune responses leads to recruitment and activation of
antigens are unlikely to represent the only immunologically myofibroblast-like cells and the progressive accumulation
productive targets, as anti-S. haematobium antibody reper- of collagen, which culminates in tissue fibrosis (80). Unlike
toire diversity beyond these specific targets remains a pow- schistosome-associated hepatic fibrosis, however, where
erful predictor of resistance (79). This observation has fibrosis has been linked to the hepatic stellate cell (30,44),
important implications for the nascent efforts to develop the origin of myofibroblast-like cells in urogenital disease
effective schistosome-specific vaccines. is unclear. Regardless, the direct biomechanical conse-
quences of fibrosis include stricture, obstruction and gen-
eral urinary dysfunction, which can progress to
IMMUNOPATHOLOGY OF UROGENITAL
obstructive renal pathology, renal failure and death
SCHISTOSOMIASIS
(81,82). Secondary consequences of chronic infection also
Schistosoma haematobium infection evokes a complex array include hematuria, increased propensity to renal and uri-
of immune responses targeting either the egg or adult nary tract coinfections and the progressive accumulation
worm/cercarial stages. As responses to the latter appear to of urothelial damage/atypia that culminates in squamous

434 © 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438
Volume 36, Number 9, September 2014 Immune responses to S. haematobium infection

cell carcinoma, an especially aggressive type of bladder demonstrated lower disease severity than uninfected con-
cancer, as well as urothelial carcinoma (83,84). trols but that this difference vanished on treatment with
In addition to the bladder pathology, 30% to 75% the schistosomicide praziquantel (105). Interestingly, func-
(depending on the population studied and examination tionally significant IL10 promoter polymorphisms corre-
techniques) of S. haematobium-infected females develop late with atopic disease and total IgE levels but not with
genital pathology detectable on examination as sandy schistosome infection susceptibility or outcome (106), sug-
patches (physical stigmata of tissue-entrapped eggs) on the gesting that IL-10 exercises a more potent influence over
cervical mucosa with the vagina, upper tract and external atopic immunopathology than schistosome-associated dis-
genitalia less commonly affected (85–90). Importantly, as ease. The mechanisms underlying this observation are
sandy patches of the cervix are pathognomonic for FGS unclear; however, a leading hypothesis involves schisto-
in women, the early appearance and subsequent evolution some-specific IgE out-competing allergen-specific IgE for
of cervical lesions in girls with FGS remains unknown. In mast cell surface receptor occupancy. It is perhaps more
addition, as the basic pathophysiology here is much the likely, however, that schistosome-elicited IL-10 directly
same as in the bladder, the tissue-specific consequences inhibits the pro-atopic activity of target lineages, as has
include increased susceptibility to sexually transmitted been demonstrated in basophils (107). As in other aspects
infections (especially HIV), dyspareunia and infertility of human schistosome studies, the significance of these
(85–90). Importantly, infection and genital disease is often effects is often unclear as effect magnitude and even direc-
established during childhood, long before the onset of sex- tion varies between reports.
ual maturity, leading to lifelong pathological consequences
(91). Less commonly, S. haematobium can involve the male
CONCLUSION
genital organs, where it generally manifests as hematosper-
mia and possibly infertility (92). Urogenital schistosomiasis comprises a significant and
Though S. haematobium infection physically localizes to under-appreciated source of human morbidity and mortal-
the urogenital tract, its infectious sequelae may systemi- ity; however, very little is known of the host or pathogen
cally manifest. Indeed, a major source of S. haematobi- determinants of disease. This lack of clarity is, in part, due
um’s disease burden manifests as developmental delay of to the relatively enigmatic nature of the aetiological agent,
chronically infected children both as a consequence of S. haematobium, and the historical lack of experimentally
nutritional deficiencies (most commonly due to iron defi- tractable disease models. With the recent advent of S. hae-
ciency associated with chronic inflammation and hematu- matobium-specific tools and a mouse model of urogenital
ria) and repeated urinary tract coinfections (93–98). infection, however, the field is poised to dissect urogenital
Moreover, S. haematobium-infected individuals demon- disease away from its better-studied hepatosplenic cousin
strate increased susceptibility to Plasmodium species due, and define the unique immunopathology that governs our
in large part, to IL-10’s influence on cytokine and anti- interaction with this deadly schistosome. To facilitate the
body responses (99,100). These same alterations in study of S. haematobium infection, we present below a
immune timbre, however, provide one of the few poten- summary of areas of particular interest to the field. Glean-
tially positive effects of infection—infected individuals ing insights in these areas will further our understanding
demonstrate substantially reduced off-target immunoreac- of fibrosing disease (both urogenital and otherwise),
tivity such as occurs in atopic and autoimmune conditions. cancer and immunopathology in general and, hopefully,
Given the potent type 2 immune deviation associated with further arm us to intervene therapeutically therein.
schistosomal infection, it is unsurprising that disease sever-
ity in such type 1-driven immune pathologies as Crohn’s
OPPORTUNITIES FOR SCHISTOSOMA
disease, type 1 diabetes and multiple sclerosis would be
HAEMATOBIUM INFECTION RESEARCH
significantly reduced (reviewed in (101)); however, similarly
beneficial effects are observed in type 2-driven pathologies • Development of animal models of S. haematobium
such as asthma and atopic disease (102,103), suggesting infection conducive to the study of the role of IgE.
that disease amelioration might be due to regulatory influ- • Identification of major secreted products of S. haemato-
ences rather than type 1-type 2 antagonism. Indeed, schis- bium eggs and comparisons of their structure and func-
tosome-infected individuals demonstrate enhanced basal tion to homologs in other schistosome species.
and stimulated IL-10 production capable of effectively • Determination of how and by what mechanisms S. hae-
inhibiting both autoimmune and atopic pathologies (104). matobium adult worms track to the pelvic circulation.
Congruently, studies of asthma in S. mansoni-endemic • Elucidation of how inflammatory cervicovaginal lesions
populations demonstrate that infected asthmatics not only evolve during female genital schistosomiasis.

© 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438 435
J. I. Odegaard & M. H. Hsieh Parasite Immunology

REFERENCES
1 World Health Organization, The Control of ings at necropsy. Am J Trop Med Hyg morbidity. Am J Trop Med Hyg 2000; 62:
Schistosomiasis. Report of a WHO Expert 1977; 26: 702–716. 19–28.
Committee. Technical Report Series no. 16 Cheever AW, Kamel IA, Elwi AM, Mosi- 29 Verwaerde C, Joseph M, Capron M, et al.
728. Geneva, WHO, 1985. mann JE, Danner R, Sippel JE. Schistoso- Functional properties of a rat monoclonal
2 Strickland GT & Ramirez BL. Schistosomi- ma mansoni and S. haematobium infections IgE antibody specific for Schistosoma
asis. In: Strickland GT, (ed): Hunters Tropi- in Egypt. III. Extrahepatic pathology. Am J mansoni. J Immunol 1987; 138: 4441–4446.
cal Medicine and Emerging Infectious Trop Med Hyg 1978; 27: 55–75. 30 Rousseaux-Prevost R, Capron M, Bazin H
Diseases. Philadelphia, W.B. Saunders Com- 17 Ghoneim I, Rabets J & Mawhorter S. Camp- & Capron A. IgE in experimental schistoso-
pany. 2000: 804–832. bell-Walsh urology/editor-in-chief, Alan J. miasis. II. Quantitative determination of
3 King CH. Health metrics for helminthic Wein; [editors, Louis R. Kavoussi… et al.]. specific IgE antibodies against S. mansoni:
infections. Adv Parasitol 2010; 73: 51–69. In: Wein AJ, Kavoussi LR & Campbell MF a follow-up study of two strains of infected
4 Gryseels B, Polman K, Clerinx J & Kestens (eds): Campbell-Walsh Urology. Philadelphia, rats. Correlation with protective immunity.
L. Human schistosomiasis. Lancet 2006; PA: Elsevier Saunders. 2011: p. 4 v. (xxxvii, Immunology 1978; 35: 33–39.
368: 1106–1118. 3753, 95 p.). 31 Gounni AS, Lamkhioued B, Ochiai K,
5 Loker ES. A comparative study of the life- 18 Sadun EH, Von Lichtenberg F, Cheever et al. High-affinity IgE receptor on eosin-
histories of mammalian schistosomes. Para- AW, Erickson DG & Hickman RL. Experi- ophils is involved in defence against para-
sitology 1983; 2: 343–369. mental infection with Schistosoma haemato- sites. Nature 1994; 367: 183–186.
6 Rheinberg CE, Mone H, Caffrey CR, bium in chimpanzees. Am J Trop Med Hyg 32 Molofsky AB, Nussbaum JC, Liang H-E,
Imbert-Establet D, Jourdane J, Ruppel A. 1970; 19: 427–458. et al. Innate lymphoid type 2 cells sustain
Schistosoma haematobium, S. intercalatum, 19 Von Lichtenberg F, Edington GM, Nwabu- visceral adipose tissue eosinophils and alter-
S. japonicum, S. mansoni, and S. rodhaini ebo I, Taylor JR & Smith JH. Pathologic natively activated macrophages. J Exp Med
in mice: relationship between patterns of effects of schistomiasis in Ibadan Western 2013; 210: 535–549.
lung migration by schistosomula and perfu- State of Nigeria. II. Pathogenesis of lesions 33 Heredia JE, Mukundan L, Chen FM, et al.
sion recovery of adult worms. Parasitol Res of the bladder and ureters. Am J Trop Med Type 2 innate signals stimulate fibro/adipo-
1998; 84: 338–342. Hyg 1971; 20: 244–254. genic progenitors to facilitate muscle regen-
7 Fu C-L, Odegaard JI, Herbert DR & Hsieh 20 Smith JH, Torky H, Kelada AS & Farid Z. eration. Cell 2013; 153: 376–388.
MH. A novel mouse model of Schistosoma Schistosomal polyposis of the urinary blad- 34 Reiman RM, Thompson RW, Feng CG,
haematobium egg-induced immunopathol- der. Am J Trop Med Hyg 1977; 26: 85–88. et al. Interleukin-5 (IL-5) augments the
ogy. PLoS Pathog 2012; 8: e1002605. 21 Wilson MS, Mentink-Kane MM, Pesce JT, progression of liver fibrosis by regulating
8 Rinaldi G, Okatcha TI, Popratiloff A, et al. Ramalingam TR, Thompson R, Wynn TA. IL-13 activity. Infect Immun 2006; 74: 1471–
Genetic manipulation of Schistosoma hae- Immunopathology of schistosomiasis. Immu- 1479.
matobium, the neglected schistosome. PLoS nol Cell Biol 2007; 85: 148–154. 35 Swartz JM, Dyer KD, Cheever AW, et al.
Negl Trop Dis 2011; 5: e1348. 22 Herbert DR, Holscher C, Mohrs M, Schistosoma mansoni infection in eosinophil
9 Young ND, Jex AR, Li B, et al. Whole- et al. Alternative macrophage activation is lineage-ablated mice. Blood 2006; 108:
genome sequence of Schistosoma haemato- essential for survival during schistosomiasis 2420–2427.
bium. Nat Genet 2012; 44: 221–225. and downmodulates T helper 1 responses 36 Phythian-Adams AT, Cook PC, Lundie
10 Beaver Jung RC & Cupp EWPC. Clinical and immunopathology. Immunity 2004; 20: RJ, et al. CD11c depletion severely
Parasitology. Philadelphia, PA, Lea and 623–635. disrupts Th2 induction and develop-
Febiger. 1984. 23 Chu D, Du M, Hu X, Wu Q & Shen J. ment in vivo. J Exp Med 2010; 207:
11 El Ridi R, Ismail S, Gaafar T & El Demel- Paeoniflorin attenuates schistosomiasis 2089–2096.
lawy M. Differential responsiveness of japonica-associated liver fibrosis through 37 Kane CM, Cervi L, Sun J, et al. Helminth
humans with early-stage schistosomiasis inhibiting alternative activation of macro- antigens modulate TLR-initiated dendritic
haematobium to Schistosoma haematobium phages. Parasitology 2011; 138: 1259–1271. cell activation. J Immunol 2004; 173: 7454–
soluble adult-worm and egg antigens. 24 Burke ML, McManus DP, Ramm GA, 7461.
Parasitol Res 1997; 83: 471–477. et al. Temporal expression of chemokines 38 Van Liempt E, van Vliet SJ, Engering A,
12 Gaafar T, Helmy M, Ismail S, Afifi A, dictates the hepatic inflammatory infiltrate et al. Schistosoma mansoni soluble egg anti-
Guirguis N, el Ridi R. Identification of the in a murine model of schistosomiasis. PLoS gens are internalized by human dendritic
Schistosoma haematobium soluble egg anti- Negl Trop Dis 2010; 4: e598. cells through multiple C-type lectins
gens inducing antibody production and/or 25 Hams E, Aviello G & Fallon PG. The and suppress TLR-induced dendritic cell
T cell proliferation in humans. J Egypt Soc Schistosoma granuloma: friend or foe? Front activation. Mol Immunol 2007; 44: 2605–
Parasitol 1992; 22: 441–451. Immunol 2013; 4: 89. 2615.
13 Gaafar T, Ismail S, Helmy M, Afifi A, 26 Ray D, Nelson TA, Fu C-L, et al. Tran- 39 Perona-Wright G, Jenkins SJ & MacDon-
Guirguis N, el Ridi R. Identification of scriptional profiling of the bladder in uro- ald AS. Dendritic cell activation and func-
Schistosoma haematobium soluble egg genital schistosomiasis reveals pathways of tion in response to Schistosoma mansoni.
antigens that elicit human granuloma inflammatory fibrosis and urothelial com- Int J Parasitol 2006; 36: 711–721.
formation in vitro. Parasitol Res 1993; 79: promise. PLoS Negl Trop Dis 2012; 6: 40 MacDonald AS, Straw AD, Dalton NM &
103–108. e1912. Pearce EJ. Cutting edge: Th2 response
14 Hagan P, Blumenthal UJ, Dunn D, Simp- 27 Eltoum IA, Suliaman SM, Ismail BM, Ismail induction by dendritic cells: a role for
son AJ & Wilkins HA. Human IgE, IgG4 AI, Ali MM, Homeida MM. Evaluation of CD40. J Immunol 2002; 168: 537–540.
and resistance to reinfection with Schistoso- eosinophiluria in the diagnosis of schistoso- 41 Blazquez AB & Berin MC. Gastrointestinal
ma haematobium. Nature 1991; 349: 243– miasis hematobium: a field-based study. Am dendritic cells promote Th2 skewing via
245. J Trop Med Hyg 1992; 46: 732–736. OX40L. J Immunol 2008; 180: 4441–4450.
15 Cheever AW, Kamel IA, Elwi AM, Mosi- 28 Reimert CM, Mshinda HM, Hatz CF, et al. 42 Jenkins SJ & Mountford AP. Dendritic cells
mann JE & Danner R. Schistosoma man- Quantitative assessment of eosinophiluria activated with products released by schisto-
soni and S. haematobium infections in in Schistosoma haematobium infections: some larvae drive Th2-type immune
Egypt. II. Quantitative parasitological find- a new marker of infection and bladder responses, which can be inhibited by

436 © 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438
Volume 36, Number 9, September 2014 Immune responses to S. haematobium infection

manipulation of CD40 costimulation. Infect worms and granuloma formation in mice chemokine receptors, CD4+CD25HIGH+
Immun 2005; 73: 395–402. infected with Schistosoma japonicum. Paras- T-cells and clinical forms of human schisto-
43 Everts B, Hussaarts L, Driessen NN, itol Res 2008; 102: 1129–1134. somiasis. Acta Trop 2008; 108: 139–149.
et al. Schistosome-derived omega-1 drives 56 Byram JE & von Lichtenberg F. Altered 69 Vella AT & Pearce EJ. CD4 + Th2
Th2 polarization by suppressing protein schistosome granuloma formation in nude response induced by Schistosoma mansoni
synthesis following internalization by the mice. Am J Trop Med Hyg 1977; 26: 944– eggs develops rapidly, through an early,
mannose receptor. J Exp Med 2012; 209: 956. transient, Th0-like stage. J Immunol 1992;
1753–1767. 57 Kouriba B, Chevillard C, Bream JH, et al. 148: 2283–2290.
44 Dunne DW, Jones FM & Doenhoff MJ. Analysis of the 5q31-q33 locus shows an 70 Isnard A & Chevillard C. Recent advances
The purification, characterization, serologi- association between IL13-1055C/T IL-13- in the characterization of genetic factors
cal activity and hepatotoxic properties of 591A/G polymorphisms and Schistosoma involved in human susceptibility to infec-
two cationic glycoproteins (alpha 1 and haematobium infections. J Immunol 2005; tion by schistosomiasis. Curr Genomics
omega 1) from Schistosoma mansoni eggs. 174: 6274–6281. 2008; 9: 290–300.
Parasitology 1991; 2: 225–236. 58 Isnard A, Kouriba B, Doumbo O & Chevil- 71 Maizels RM. Parasite immunomodulation
45 Remick DG, Chensue SW, Hiserodt JC, lard C. Association of rs7719175, located in and polymorphisms of the immune system.
Higashi GI & Kunkel SL. Flow-cytometric the IL13 gene promoter, with Schistosoma J Biol 2009; 8: 62.
evaluation of lymphocyte subpopulations in haematobium infection levels and identifica- 72 Mallevaey T, Fontaine J, Breuilh L, et al.
synchronously developing Schistosoma man- tion of a susceptibility haplotype. Genes Im- Invariant and noninvariant natural killer T
soni egg and Sephadex bead pulmonary mun 2011; 12: 31–39. cells exert opposite regulatory functions on
granulomas. Amer J Pathol 1988; 131: 298– 59 He H, Isnard A, Kouriba B, et al. A the immune response during murine
307. STAT6 gene polymorphism is associated schistosomiasis. Infect Immun 2007; 75:
46 Asseman C, Pancre V, Quatennens B & with high infection levels in urinary schisto- 2171–2180.
Auriault C. Schistosoma mansoni-infected somiasis. Genes Immun 2008; 9: 195–206. 73 Mallevaey T, Zanetta JP, Faveeuw C, et al.
mice show augmented hepatic fibrosis and 60 Yap G, Cheever A, Caspar P, Jankovic D & Activation of invariant NKT cells by the
selective inhibition of liver cytokine produc- Sher A. Unimpaired down-modulation of helminth parasite Schistosoma mansoni. J
tion after treatment with anti-NK1.1 anti- the hepatic granulomatous response in CD8 Immunol 2006; 176: 2476–2485.
bodies. Immunol Lett 1996; 54: 11–20. T-cell- and gamma interferon-deficient mice 74 Ji F, Liu Z, Cao J, Li N, Liu Z, et al. B cell
47 Hou X, Yu F, Man S, et al. Negative regula- chronically infected with Schistosoma man- response is required for granuloma forma-
tion of Schistosoma japonicum egg-induced soni. Infect Immun 1997; 65: 2583–2586. tion in the early infection of Schistosoma
liver fibrosis by natural killer cells. PLoS Negl 61 Chensue SW, Warmington K, Ruth JH, japonicum. PLoS ONE 2008; 3: e1724.
Trop Dis 2012; 6: e1456. Lukacs N & Kunkel SL. Mycobacterial 75 Nash TE. Antibody response to a polysac-
48 Hashimoto A, Pincelli C, Fujioka A, and schistosomal antigen-elicited granu- charide antigen present in the schistosome
Fukuyama K & Epstein WL. Relationship loma formation in IFN-gamma and IL-4 gut. I. Sensitivity and specificity. Am J Trop
between NK cells and granulomatous knockout mice: analysis of local and regio- Med Hyg 1978; 27: 939–943.
inflammation in mice. J Clin Lab Immunol nal cytokine and chemokine networks. J 76 Grogan JL, Kremsner PG, van Dam GJ, De-
1990; 33: 41–47. Immunol 1997; 159: 3565–3573. elder AM & Yazdanbakhsh M. Anti-schisto-
49 Gao B & Radaeva S. Natural killer and 62 Wynn TA, Morawetz R, Scharton-Kersten some IgG4 and IgE at 2 years after
natural killer T cells in liver fibrosis. T, et al. Analysis of granuloma formation in chemotherapy: infected versus uninfected
Biochim Biophys Acta 2013; 1832: 1061– double cytokine-deficient mice reveals a cen- individuals. J Infect Dis 1997; 176: 1344–
1069. tral role for IL-10 in polarizing both T 1350.
50 Niel G, Sainte-Laudy J, Carme B, et al. The helper cell 1- and T helper cell 2-type cyto- 77 Iskander R, Das PK & Aalberse RC. IgG4
comparative value of the human basophil kine responses in vivo. J Immunol 1997; 159: antibodies in Egyptian patients with schis-
degranulation test and classical serology 5014–5023. tosomiasis. Int Arch Allergy Immunol 1981;
(immunofluorescence, haemagglutination, 63 Burke ML, Jones MK, Gobert GN, Li YS, 66: 200–207.
electrosyneresis, Vogel and Minnings Test) in Ellis MK, McManus DP. Immunopathogene- 78 Naus CW, van Dam GJ, Kremsner PG,
the diagnosis of bilharzioses due to Schisto- sis of human schistosomiasis. Parasite Immu- Krijger FW & Deelder AM. Human IgE,
soma mansoni and Schistosoma haematob. nol 2009; 31: 163–176. IgG subclass, and IgM responses to worm
Ann Biol Clin 1982; 40: 573–577. 64 Wahl SM, Frazier-Jessen M, Jin WW, Kopp and egg antigens in schistosomiasis haemat-
51 Schramm G, Mohrs K, Wodrich M, et al. JB, Sher A, Cheever AW. Cytokine regula- obium: a 12-month study of reinfection in
Cutting edge: IPSE/alpha-1, a glycoprotein tion of schistosome-induced granuloma and Cameroonian children. Clin Infect Dis
from Schistosoma mansoni eggs, induces IgE- fibrosis. Kidney Int 1997; 51: 1370–1375. 1998; 26: 1142–1147.
dependent, antigen-independent IL-4 produc- 65 Okano M, Satoskar AR, Nishizaki K, Abe 79 Mutapi F, Burchmore R, Mduluza T, Midzi
tion by murine basophils in vivo. J Immunol M & Harn DA. Induction of Th2 responses N, Turner CMR, Maizels RM. Age-related
2007; 178: 6023–6027. and IgE is largely due to carbohydrates and infection intensity-related shifts in anti-
52 Sullivan BM, Liang H-E, Bando JK, et al. functioning as adjuvants on Schistosoma body recognition of defined protein anti-
Genetic analysis of basophil function in mansoni egg antigens. J Immunol 1999; 163: gens in a schistosome-exposed population.
vivo. Nat Immunol 2011; 12: 527–535. 6712–6717. J Infect Dis 2008; 198: 167–175.
53 Ganley-Leal LM, Mwinzi PNM, Cetre-Sos- 66 Lundy SK & Boros DL. Fas ligand-express- 80 Rauterberg J, Voss B, Pott G & Gerlach U.
sah CB, et al. Higher percentages of circu- ing B-1a lymphocytes mediate CD4(+)-T-cell Connective tissue components of the nor-
lating mast cell precursors correlate with apoptosis during schistosomal infection: mal and fibrotic liver. I. Structure, local dis-
susceptibility to reinfection with Schistoso- induction by interleukin 4 (IL-4) and IL-10. tribution and metabolism of connective
ma mansoni. Am J Trop Med Hyg 2006; 75: Infect Immun 2002; 70: 812–819. tissue components in the normal liver and
1053–1057. 67 Reis EAG, Azevedo TM, McBride AJA, changes in chronic liver diseases. Klinische
54 Chan CY, St John AL, Abraham SN & St. Harn DA & Reis MG. Na€ıve donor Wochenschrift 1981; 59: 767–779.
John AL. Mast cell interleukin-10 drives responses to Schistosoma mansoni soluble 81 Von Lichtenberg F. Schistosomiasis
localized tolerance in chronic bladder infec- egg antigens. Scand J Immunol 2007; 66: as a worldwide problem: pathology. J Toxi-
tion. Immunity 2013; 38: 1–11. 662–670. col Environ Health 1975; 1: 175–184.
55 Cheng Y, Song W, Liu W, et al. The effects 68 Teixeira-Carvalho A, Martins-Filho OA, 82 Abdel-Wahab MF, Ramzy I, Esmat G, El
of T cell deficiency on the development of Peruhype-Magalh~aes V, et al. Cytokines, Kafass H & Strickland GT. Ultrasound for

© 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438 437
J. I. Odegaard & M. H. Hsieh Parasite Immunology

detecting Schistosoma haematobium urinary 91 Hegertun IEA, Sulheim Gundersen KM, 100 Diallo TO, Remoue F, Schacht AM, et al.
tract complications: comparison with radio- Kleppa E, et al. S. haematobium as a com- Schistosomiasis co-infection in humans
graphic procedures. J Urol 1992; 148: mon cause of genital morbidity in girls: a influences inflammatory markers in uncom-
346–350. cross-sectional study of children in South plicated Plasmodium falciparum malaria.
83 Gelfand M, Weinberg RW & Castle WM. Africa. PLoS Negl Trop Dis 2013; 7: e2104. Parasite Immunol 2004; 26: 365–369.
Relation between carcinoma of the bladder 92 Corachan M, Valls ME, Gascon J, Almeda 101 Cardoso LS, Oliveira SC & Araujo MI.
and infestation with Schistosoma haemato- J & Vilana R. Hematospermia: a new etiol- Schistosoma mansoni antigens as modula-
bium. Lancet 1967; 1: 1249–1251. ogy of clinical interest. Am J Trop Med tors of the allergic inflammatory response
84 Brand KG. Schistosomiasis--cancer: etiolog- Hyg 1994; 50: 580–584. in asthma. Endocr Metab Immune Disord
ical considerations. A review. Acta Trop 93 Farid Bassily S, Schulert AR, Zeind AS, Drug Targets 2012; 12: 24–32.
1979; 36: 203–214. McConnell E, Abdel Wahab MF. Urinary 102 Medeiros M, Figueiredo JP, Almeida MC,
85 Kjetland EF, Kurewa EN, Ndhlovu PD, blood loss in Schistosoma haematobium et al. Schistosoma mansoni infection is asso-
et al. Female genital schistosomiasis--a dif- infection in Egyptian farmers. Trans R Soc ciated with a reduced course of asthma. J
ferential diagnosis to sexually transmitted Trop Med Hyg 1968; 62: 496–500. Allergy Clin Immunol 2003; 111: 947–951.
disease: genital itch and vaginal discharge as 94 Tatala SR, Kihamia CM, Kyungu LH & 103 Araujo MI, Lopes AA, Medeiros M, et al.
indicators of genital Schistosoma haematobi- Svanberg U. Risk factors for anaemia in Inverse association between skin response
um morbidity in a cross-sectional study in schoolchildren in Tanga Region, Tanzania. to aeroallergens and Schistosoma mansoni
endemic rural Zimbabwe. Trop Med Int Tanzan J Health Res 2008; 10: 189–202. infection. Int Arch Allergy Immunol 2000;
Health 2008; 13: 1509–1517. 95 The Partnership for Child Development. 123: 145–148.
86 Jourdan PM, Roald B, Poggensee G, Gun- The health and nutritional status of school- 104 Ara ujo MI, Hoppe BS, Medeiros M &
dersen SG & Kjetland EF. Increased vascu- children in Africa: evidence from school- Carvalho EM. Schistosoma mansoni infec-
larity in cervicovaginal mucosa with based health programmes in Ghana and tion modulates the immune response
Schistosoma haematobium infection. PLoS Tanzania. The Partnership for Child Devel- against allergic and auto-immune diseases.
Negl Trop Dis 2011; 5: e1170. opment. Trans R Soc Trop Med Hyg 1998; Mem Inst Oswaldo Cruz 2004; 99: 27–32.
87 Kjetland EF, Mduluza T, Ndhlovu PD, 92: 254–261. 105 Lynch NR, Hagel I, Perez M, Di Prisco MC,
et al. Genital schistosomiasis in women: 96 Nmorsi OP, Kwandu UN & Ebiaguanye Lopez R, Alvarez N. Effect of anthelmintic
a clinical 12-month in vivo study follow- LM. Schistosoma haematobium and urinary treatment on the allergic reactivity of chil-
ing treatment with praziquantel. tract pathogens co-infections in a rural dren in a tropical slum. Current Genomics
Trans R Soc Trop Med Hyg 2006; 100: community of Edo State, Nigeria. J Com- 1993; 9: 290–300.
740–752. mun Dis 2007; 39: 85–90. 106 Grant AV, Araujo MI, Ponte EV, et al.
88 Kjetland EF, Leutscher PDC & Ndhlovu 97 Kassim OO & Stek M Jr. Bacteriuria and Polymorphisms in IL10 are associated with
PD. A review of female genital schistosomi- hematuria in infections due to Schistosoma total Immunoglobulin E levels and Schisto-
asis. Trends Parasitol 2012; 28: 59–66. haematobium. J Infect Dis 1983; 147: 960. soma mansoni infection intensity in a Bra-
89 Kjetland EF, Ndhlovu PD, Mduluza T, 98 Wilkins HA. Schistosoma haematobium in a zilian population. Genes Immun 2011; 12:
et al. Simple clinical manifestations of geni- Gambian community. III. The prevalence 46–50.
tal Schistosoma haematobium infection in of bacteriuria and of hypertension. 107 Larson D, H€ ubner MP, Torrero MN, et al.
rural Zimbabwean women. Am J Trop Ann Trop Med Parasitol 1977; 71: 179–186. Chronic helminth infection reduces basophil
Med Hyg 2005; 72: 311–319. 99 Courtin D, Djilali-Sa€ıah A, Milet J, et al. responsiveness in an IL-10-dependent
90 Helling-Giese G, Sjaastad A, Poggensee G, Schistosoma haematobium infection affects manner. J Immunol 2012; 188: 4188–4199.
et al. Female genital schistosomiasis (FGS): Plasmodium falciparum-specific IgG
relationship between gynecological and his- responses associated with protection against
topathological findings. Acta Trop 1996; 62: malaria. Parasite Immunol 2011; 33: 124–
257–267. 131.

438 © 2013 John Wiley & Sons Ltd, Parasite Immunology, 36, 428–438

You might also like