Vous êtes sur la page 1sur 11

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/230862981

Virulence factors of schistosomes

Article in Microbes and Infection September 2012


DOI: 10.1016/j.micinf.2012.09.001 Source: PubMed

CITATIONS READS

10 399

1 author:

Robert Alan Wilson


The University of York
297 PUBLICATIONS 8,324 CITATIONS

SEE PROFILE

All in-text references underlined in blue are linked to publications on ResearchGate, Available from: Robert Alan Wilson
letting you access and read them immediately. Retrieved on: 02 October 2016
This article appeared in a journal published by Elsevier. The attached
copy is furnished to the author for internal non-commercial research
and education use, including for instruction at the authors institution
and sharing with colleagues.
Other uses, including reproduction and distribution, or selling or
licensing copies, or posting to personal, institutional or third party
websites are prohibited.
In most cases authors are permitted to post their version of the
article (e.g. in Word or Tex form) to their personal website or
institutional repository. Authors requiring further information
regarding Elseviers archiving and manuscript policies are
encouraged to visit:
http://www.elsevier.com/copyright
Author's personal copy

Microbes and Infection 14 (2012) 1442e1450


www.elsevier.com/locate/micinf

Virulence factors of schistosomes


R. Alan Wilson*
Centre for Immunology & Infection, Department of Biology, University of York, Heslington, York YO10 5DD, UK

Received 17 April 2012; accepted 3 September 2012


Available online 11 September 2012

Abstract

This review considers whether the products of schistosomes in the mammalian host can be considered as virulence factors. These include: the
cercarial secretions used in infection, those of the migrating schistosomulum, surface-exposed proteins of adult worms in the portal system and
their gut vomitus in the context of immune evasion, secretions of the egg facilitating its escape from gut tissues and micro-exon gene products.
2012 Institut Pasteur. Published by Elsevier Masson SAS. All rights reserved.

Keywords: Schistosoma mansoni; Tegument; Host physiology; MEGs

1. Introduction with sequencing of the genomes and transcriptomes of the


major schistosome species have made it possible, for the first
The concept of virulence factors first came to prominence time, to define the major constituents of the schistoso-
in the 1970s, applied almost exclusively to pathogenic meemammalian host interface, and the secretions of the larval
bacteria. The term encompasses microbial products, generally and adult stages associated with the host tissues and blood-
macromolecules, which facilitate establishment and mainte- stream [2]. Can these constituents be termed or represented as
nance of an infection, contributing to its pathogenicity. They virulence factors?
include cell surface components essential for infection and
evasion of the immune response, as well as secreted toxins that 2. Schistosomes and the mammalian host
are directly linked to virulence. The purpose of this article is to
evaluate whether the virulence factor concept is applicable to The schistosomes parasitic in humans are threadlike and
macroparasitic helminths, specifically the blood dwelling about 1 cm long, the adults living as paired males and females
schistosomes of humans. It has only been used overtly with the in the hepatic portal vasculature (S. mansoni and Schistosoma
helminth parasite Schistosoma mansoni in a single paper japonicum) or the vessels of the bladder wall (Schistosoma
published in 2011 [1]. The task is not straightforward because, haematobium). The eggs laid by the females in the tissues of
apart from scale, there is a major difference between microbes the gut or bladder wall are the main pathogenic agents. In the
and helminths. Once a microbe has established it undergoes intestinal infections they may dislodge and travel downstream
replication, usually rapid, until it is checked by the immune to embolise in the liver; in the case of S. haematobium they
system or the host is overwhelmed. Conversely, helminths do have a direct effect on the tissues of the urogenital tract. The
not multiply in the mammalian host, each invasion being adult worms are remarkable for living, apparently unprotected,
a singular event. Thus, the parasite burden accumulates and in the host bloodstream for decades so must clearly deploy
may be terminated acutely or persist chronically according to sophisticated means of immune evasion. The infective agents
helminth species. The recent advances in proteomics, coupled are microscopic cercaria larvae, shed from fresh water snail
intermediate hosts, which directly penetrate the skin to reach
dermal blood vessels and begin migration to the portal system.
* Tel.: 44 1904 328600; fax: 44 1904 328505. The dramatic physiological transition from fresh water to the
E-mail address: raw3@york.ac.uk. human body is accompanied by loss of the cercarial tail and

1286-4579/$ - see front matter 2012 Institut Pasteur. Published by Elsevier Masson SAS. All rights reserved.
http://dx.doi.org/10.1016/j.micinf.2012.09.001
Author's personal copy

R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450 1443

metamorphosis into the schistosomulum. Coincident with this, Furthermore, a microarray analysis of the infection process
the parasite replaces its surface membranes and becomes indicates that five of them were the most highly-upregulated
invulnerable to killing by antibody-dependent killing mecha- metalloprotease genes in the germ ball stage, when cercarial
nisms (ADCC) [3]. The cercaria possesses five pairs of proteins are being synthesised [8]. Given the role of
acetabular glands to assist skin penetration, and a minute head mammalian metalloproteases in degradation of tissues
gland that facilitates parasite entry into a blood vessel. It is matrices, a thorough investigation of these enzymes in the
apparent that the schistosome eggs, deposited in host tissues, context of skin invasion is warranted. Why up to seven elas-
have their own system for secreting proteins required for exit tases and five metalloproteases, representing 34% and 13%
from the host. Production of factors essential for onward respectively of the material released from the pre-acetabular
transmission is not an aspect of virulence that has been applied glands, are needed for skin invasion is unclear, unless they
to microbes but it seems apposite for schistosomes. have very fine distinctions in substrate specificity. It has been
suggested that expressing multiple copies of genes for abun-
3. Host invasion dant secretory proteins might itself be part of an evasion
strategy [8]. It is noteworthy that skin penetration by cercariae
We are on safe ground in suggesting that the secretions of Trichobilharzia regenti (a bird schistosome) and S. japo-
used by cercariae to penetrate host skin are directly analogous nicum appears to be facilitated by a cysteine peptidase,
to bacterial virulence factors. However, while the work on cathepsin B2, so there may be some latitude in the enzymes
schistosomes has focused largely on proteases, with little involved in establishment among different schistosomes
information on adhesion molecules, the situation is reversed in [11,12]. Finally, experiments in vitro suggest that the serine
bacteria with the emphasis placed on adhesion proteins. proteases secreted by cercariae can cleave host immunoglob-
Additionally, bacteria secrete a whole range of exotoxins; ulins such as IgE [13] and interfere with complement-
schistosomes may produce the equivalent, but data on their mediated lysis [14]; such properties deployed in vivo could
function is lacking at present. The three pairs of cercarial post- well enhance parasite establishment in the host.
acetabular glands were shown decades ago to produce mucins It is possible that the acetabular gland secretions also
[4] that enabled the cercaria to make firm adhesions to the contain the schistosome equivalents of bacterial toxins. Cur-
stratum corneum of the skin, preparatory to penetration. wen et al. [9], identified three proteins with SCP (sperm coat
However, no attempt to date appears to have been made to protein) domains that have since been classified as part of
characterise their molecular composition. Instead, researchers a 28-member family of VAL (venom allergen like) proteins,
have focused on the soluble proteins packaged in vesicles in approximately two thirds of which are secreted by different
the two pairs of pre-acetabular glands. stages. The three constituents of the cercarial secretions are
The soluble serine proteases of S. mansoni cercariae have SmVAL4, SmVAL10 and SmVAL18, collectively representing
been well characterised over the last 20 years. They are 3% of released protein. Similar proteins have been identified
generally referred to as elastases [5] but this has provoked in other helminth secretions, e.g. Heligmosomoides polygyrus
controversy because the acetabular gland cells where they are [15]. The problem is that no clear function has yet emerged for
synthesised have disappeared before the vast majority of any VAL in any helminth species. The same is true of another
schistosomula enter the dermis where elastin fibres are situ- enigmatic constituent of the cercarial secretions, Sm16
ated [6]. At least three of these serine proteases have been (SmSLP/SPO-1), originally ascribed anti-inflammatory prop-
biochemically characterised from the acetabular gland secre- erties [16], and more recently a caspase-dependent apoptotic
tions [7] and their transcripts identified in the precursor germ function [17]. Its presence in extruded material may simply
balls of the daughter sporocyst stage in the snail host [8]. (The reflect the fact that acetabular glands release their contents by
latest version (v5) of the S. mansoni genome lists seven holocrine secretion. Indeed, the localisation of Sm16 tran-
homologues but gives no information on the stage where scripts to numerous internal tissues of the cercaria by
each is expressed.) Since the majority of S. mansoni parasites WISH [18], plus the absence of a signal peptide suggests that
do not enter the dermis for at least 48 h after penetration, it has an internal function in the newly penetrated parasite.
the substrate of the characterised serine proteases is likely Potentially, if it is pro-apoptotic, this could be in the
to be the proteins of the stratum corneum and the epidermis tissue remodelling that accompanies transformation to the
where the incoming cercarial body creates a penetration tun- schistosomulum.
nel (http://www.york.ac.uk/cii/media-library/cercariae/). The The final constituents of cercarial secretions that merit
cercarial tail and the thick protective glycocalyx are shed as attention as virulence factors are the glycans. Both the proteins
the parasite enters the epidermis. in soluble cercarial secretions and those in the soluble egg
A proteomic analysis of the soluble secretions released by secretions are heavily glycosylated [19,20]. A plethora of both
the schistosome cercariae during artificial transformation N- and O-linked glycan structures have been identified by
in vitro [9] identified a hitherto unsuspected major component mass spectrometry and many proved to be shared between the
as a metalloprotease. This is now classified in the S. mansoni two transmission stages, suggesting a commonality of function
genome database as an invadolysin (Smp_090100). Whilst by the stages entering and leaving the host [19,20]. Moreover,
tandem mass spectrometry pinpointed only a single protein, v5 these glycans are highly immunogenic [21,22], but there is no
of the S. mansoni genome lists seven homologues [10]. evidence that the responses invoked are protective. This has
Author's personal copy

1444 R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450

led to the suggestion that they are part of the immune evasion macrophages and functionally compromised MHCIIhi cells
mechanism, serving as a smoke screen to deflect the antibodies that are unable to present antigen.
away from vulnerable and functional peptide epitopes [21,23]
or in the matadors cloak variant of the hypothesis [20], actu- 4. Intravascular migration
ally luring leucocytes away from the incoming larva so that it
can escape attack. Much less is known about the factors secreted by the
The molecular definition of secreted factors released by schistosomulum after it has completed its transformation in
invading schistosomes is ahead of the research to delineate the skin, shedding its cercarial plasma membrane and
their functions. Nevertheless, evidence is accumulating for replacing it with a new surface in an apparently seamless
their interaction with the dermal immune response, although process. (Whether that shed material represents a source of
the identity of the specific factors involved has yet to be signals to the host skin has not been investigated.) The
elucidated. Since the process of schistosome infection, per se, parasites use of the head gland to gain access to a blood vessel
does not appear to elicit protective immunity, it is reasonable was documented in ultrastructural studies [27], but its minute
to assume that observed effects are parasite-, not host- size means that little material is available for analysis.
protective. The secretions from CFDA-SE (carboxy- Biosynthetic labelling with 35S-methionine has revealed that
fluorescein diacetate-succinimidyl ester) labelled cercariae skin and lung stage schistosomula release a dominant protein
released into the skin during penetration are initially taken up of wMr 20 kDa into the external environment [28]. In 2010 it
by neutrophils [24]. By 24 h dermal macrophages and was identified by proteomics as a mixture of two proteins,
dendritic cells are labelled, and both of these cells are MEG-3.1 and MEG-3.2, encoded by micro-exon genes ([29]
observed in the skin-draining lymph nodes at 48 h, perhaps see Egg escape). Furthermore, whole mount in situ hybrid-
a reflection of their transit time from the skin. In vitro, isation (WISH) revealed that expression of the MEG-3.2 gene
macrophages are slower to take up the secretions than was localised to the head gland [29], (the only transcript so far
dendritic cells but produce more (suppressive) IL-10 [24]. identified from that tissue) and to a lesser extent the tegument
Ligands for the mannose receptor (CD206), presumably cells bodies (Fig. 1A; Parker-Manuel, pers. comm.). This
glycans, are abundant in the secretions [25]. In this context it indicates that the MEG-3 proteins are likely to be released
is notable that schistosomes, unlike the mammalian hosts, do across the tegument surface as well as from the head gland and
not terminate their sugar chains with sialic acid, lacking the makes them candidate constituents of the enigmatic homoge-
necessary transferase enzyme. Furthermore, in mannose neous secretory bodies, uniquely found in the lung stage [30].
receptor-null mice, after exposure to cercariae, fewer leuco- For information on other likely secreted or exposed proteins of
cytes in the skin had taken up parasite antigen but there was migrating lung schistosomula we have to rely on gene
enhanced proliferation of the skin-draining lymph node cells expression patterns revealed by cDNA sequencing or micro-
secreting interferon gamma. Thus it appears that the cercarial array analyses, which have identified known tegument
secretions down-modulate the production of pro-inflammatory constituents of the adult worm [31].
cytokines in the skin. This conclusion is reinforced by the
observation that multiple exposures of mice to cercariae 5. The life of adult worms in the bloodstream
(before the onset of egg production) induce hypo-
responsiveness of CD4 T cells in the skin-draining lymph In most hosts once migrating schistosomes have reached
nodes [26]. The crucial cells are alternatively activated the hepatic portal blood vessels and begun to feed on

Fig. 1. A, Whole mount in situ hybridisation (WISH) of the MEG-3.2 gene in a day-3 schistosomulum. The dark staining area at the anterior reveals a high level of
expression in the head gland. The lighter areas are tegument cell bodies. B, A mature egg with the sub-shell envelope syncytium highlighted by fluorescent dextran;
an unstained miracidium (m) is visible in the centre. C, In situ hybridisation to detect MEG-3.2 expression performed on liver sections containing mature eggs. The
mRNA revealed by the blue stain is localised to the sub-shell envelope.
Author's personal copy

R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450 1445

erythrocytes, they appear to be invulnerable to immune attack; induction of tolerance to schistosome antigens in the liver is
the exceptions are the laboratory rat and the rhesus macaque, clearly warranted.
which eliminate pre-adult and adult worms, respectively [32].
5.2. The tegument and immune evasion
5.1. Products of the schistosome gut
The schistosome interface with the bloodstream, termed
The schistosome gut is tidal, with food entering and waste a tegument, is a syncytial layer of cytoplasm, connected to cell
leaving via the mouth. Essentially it is an elongate tube bodies beneath the musculature. The unusual appearance of its
(in some places bifurcated) comprising a uniform layer of surface in juvenile and adult schistosomes, when stabilised by
epithelium, sometimes referred to as the gastrodermis. This uranyl acetate, was described w40 years ago as a heptalami-
tube is invested by smooth muscle fibres responsible for the nate membrane [30], and this term is still used by some
peristalsis that propels the contents along the lumen. The researchers. However, the structure was interpreted as
females of the species are more voracious than the males, a normal plasma membrane overlain by a laminate secretion,
consuming eight times as much blood [33]. Haemoglobin from the membranocalyx, which originated in multilaminate vesi-
ingested erythrocytes is the major protein source but plasma cles present within the cytosol of the tegument syncytium [41].
proteins, especially albumin, are also important [34]. A black It was suggested that the secreted layer lacked macromolec-
haematin/haemozoin pigment, the residue of haemoglobin ular components and so acted as a barrier to prevent antibodies
digestion is periodically voided into the portal blood and then from binding to vulnerable functional proteins in the plasma
filtered out by the resident macrophages of the liver. As membrane. This hypothesis was based on two of observations.
a detoxification product it is highly likely that the haemozoin Firstly, binding of anti-worm antibodies to the tegument
is relatively inert. Indeed the version from S. japonicum failed surface of live worms cannot easily be demonstrated, so
to exert any significant effects on the development and activ- something must be getting in the way. Secondly, without
ities of murine myeloid dendritic cells [35]. Other major uranyl acetate treatment the processing of worms for electron
products of the gut vomitus are the circulating anodic and microscopy with organic solvents extracts most of the vesicle
cathodic polysaccharide antigens, which were shown to contents, revealing their predominantly lipid composition, and
comprise the glycocalyx lining the gut [36] and subsequently the membranocalyx disappears. However, a small residue
developed as the basis for diagnostic tests [37]. No evidence remained at the centre of each empty vesicle, which stained
has been presented for effects on host physiology other than positively for glycans [41]. Furthermore, incubation of live
the formation of immune complexes with antibodies that worms with the lectin Concavalin A froze vesicles in the act
facilitates antigen clearance. Finally, as a result of earlier of fusion with the plasma membrane, revealing the presence of
biochemical studies and recent proteomic analyses we now exposed mannose residues in the membranocalyx before
have information on the proteins that are secreted into the secretion [42]. Recent proteomic analyses indicate that the
worm gut, and ultimately voided into the bloodstream [34,38]. hypothesis of the membranocalyx as an immunologically inert
These come in three categories, hydrolytic enzymes, carrier barrier might need to be modified.
proteins, and protease inhibitors, many of which are associated Clearly, the tegument surface is the principal location
with lysosomal activity. The enzymes include Cathepsins B1 where evasion of the host immune response occurs. Its prop-
(2 isoforms), C and K/S, a Pro-X carboxylpeptidase and erties and composition have been comprehensively reviewed
a Glucan 1,4 beta-glucosidase. The transporters include in recent years [43e45] and proteomics has contributed much
Ferritins, Saposins, NPC-like cholesterol binding protein, and of the new knowledge about the surface layers [2]. However,
Calumenin, a Ca transporter. The protease inhibitors identified some misguided conclusions have been reached by researchers
are a serpin and the schistosome homologue of alpha-2- who confused the secretome with the necrotome (the
macroglubulin. contaminating cytosolic proteins released by damaged or
Since adult intestinal schistosomes inhabit the hepatic dying parasites). The most exposed proteins have been tagged
portal venules of the gut wall, the contents of their guts that are by biotinylation of live worms [46,47] while GPI-anchored
voided into the bloodstream will be carried downstream to the proteins have been detached by incubation with phosphatidyl
liver. The role of the liver as an organ of the immune system inositol phospholipase C (PiPLC). In the context of virulence
[39], particularly in the context of tolerance generation [40], is factors there are three aspects to be considered. Firstly, the
now well established. Therefore, it seems surprising that there actual composition of the membranocalyx lipid barrier, and of
is little information on the interaction of any of the protein the endogenous glycans associated with lipids or proteins, is
constituents of gut vomitus with hepatic antigen presenting completely unexplored. Secondly, the proteins exposed on the
cells. Of course, this might be due to the difficulty of stimu- surface may function in a parasite-protective role. Thirdly, any
lating worms in vitro to open their mouths and regurgitate their proteins and/or glycan constituents of the membranocalyx,
gut contents [34]. However, in vivo the gut vomitus must flow released when it turns over into the bloodstream, could serve
past the fixed antigen presenting cells of the sinusoids. Hae- as modifiers of host responses or physiology.
mozoin pigment is very evident in the resident Kupffer cells of A small number of enzymes exposed on the outer leaflet of
the sinusoids and its presence implies that other vomitus the tegument plasma membrane have been implicated as
constituents will also be endocytosed; an investigation of the potential virulence factors due to their ability to modify the
Author's personal copy

1446 R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450

immediate environment of the parasite in the blood vessel a protective effect in graft-v-host disease [56]; does it modify
(reviewed by Bhardwaj & Skelly [48]). The demonstration of the interaction of host leucocytes with the tegument surface?
ATP-diphosphohydrolase activity externally on the tegument
led to the suggestion that the enzyme could regulate the 6. Egg escape from host tissues
concentration of purine nucleotides around the parasites and
hence enable them to escape the host haemostasis by pre- The schistosome egg is a composite structure comprising
venting ADP-induced platelet activation [49]. The GPI- w30 vitelline cells and a fertilised ovum packaged within an
anchored ADP-ribosyl cyclase enzyme also on the external elastic tanned protein shell, laid by a female in an undeveloped
surface, whilst apparently unable to generate cyclic adenosine state in a venule. The ovum develops into a ciliated mira-
diphosphate ribose, may catabolize extracellular NAD() to cidium whilst the egg remains in the vessel using nutrients
prevent its use by host enzymes that facilitate immune from the vitelline cells but also, since it has been estimated to
responses [50]. It has also been suggested that the adenosine increase in mass by 3.33, acquiring them from the host as
generated from adenosine monophosphate by tegumental well [57]. During development 3e4 cells detach from the
alkaline phosphatase acts as a localised immunosuppressant embryonic mass and move to the periphery where they fuse to
[51]. Lastly, the tegumental phosphodiesterase, SmNPP-5, has form the syncytial sub-shell envelope (Fig. 1B), which
actually been directly implicated as a virulence factor by the differentiates into a protein synthesising and exporting tissue
use of RNAi treatment to suppress gene expression in para- [57]. However, secretion only becomes appreciable when the
sites, followed by demonstration of their greatly reduced miracidium is mature, clearly a device to prevent premature
ability to establish after administration to mice [1]. expulsion of the partially developed egg. Release of secretions
Although many tegument surface constituents have been is the trigger that initiates egg migration across the tissues to
identified, the difficulty is to place them geographically within the gut (or bladder) lumen, and voiding in excreta. The role of
the complex surface structure. Currently, six proteins: Sm200, these secretions from live eggs is thus crucial for parasite
Sm29, Sm13, Annexin Smp_077720, Tetraspanin-TSP2, and transmission and should not be confused with the products of
Low Molecular Weight Protein (LMWP) are the prime candi- moribund or dead eggs (not considered here) that contribute to
dates for location in the secreted membranocalyx [2]; since the granulomatous pathology that is the hallmark of the disease.
membranocalyx turns over into the external environment [42], Definition of the proteins secreted by viable mature eggs to
this means they will be released into the bloodstream. Inde- facilitate escape from host tissues has proved controversial. The
pendent evidence for this fate comes in part from short term first report detailed a large number of intracellular proteins
culture experiments to collect vomitus (and inevitably any true identified by tandem mass spectrometry and postulated their
surface secretions) [34] that identified Sm200. In addition, this release via a non-classical pathway [58]. A later report
protein has been reported from the lipid fraction of host serum, described a very different situation with a relatively simple
providing confirmation of its release [52]. What the function of mixture secreted from eggs that were judged to be >96% viable
these proteins might be is a matter of conjecture, but it seems [59]. The secretions were dominated by previously described
likely that the annexin and tetraspanin play a structural role; that IPSE (Interleukin-4 inducing principle of schistosome eggs
leaves Sm200, Sm29, Sm13 and LMWP as potential virulence [60];) and Omega-1 [61], plus a few minor spots When the
factors interacting with the host. secretions were depleted of the heavily glycosylated IPSE and
The apparent invulnerability of the adult worm to antibody Omega-1 using Aleuria aurantia agglutinin the minor spots
binding and immune attack was noted above. It therefore came were greatly enriched and many could be identified by mass
as a surprise that compositional analysis of the tegument spectrometry [29]. The bulk of the minor components were
surface complex identified the presence of host antibodies of encoded by micro-exon genes, five from the MEG-2 and three
IgM, IgG1 and IgG3 classes [53,54]. Furthermore the C3 and from the MEG-3 family, with at least 10 variants of the former
C4 components of the complement cascade were also identi- and 12 of the latter. IPSE was demonstrated by immunocyto-
fied but not those of the membrane attack complex C5eC9. chemistry to originate in the sub-shell envelope [60] whilst
This implies that complement fixation is initiated but then MEG-3.2 gene was revealed by WISH to be transcribed in the
inhibited at an early stage. Possible candidates for this role are same tissue (Fig. 1C). As a footnote, the recently characterised
the two GPI-anchored schistosome homologues of host CD59 Sm-kappa antigenic glycoprotein [62] is associated with the
(Smp_019350, Smp_105220), that can be detached from live miracidium and not found in the egg secretions [59,62],
worms by PiPLC treatment [31]. The human equivalent providing strong evidence that the sub-shell envelope is the true
protects self-cells by binding to the membrane attack complex. interface between the living egg and the host tissues. The glycan
Counter to this suggestion, a quantitative estimate of the composition of the egg secretions has been characterised,
amounts of C3 and IgG at the schistosome surface using the revealing a wide variety of N- and O-linked structures, many of
QconCAT technique [55] found much less C3, indicating that them shared with the secretions of the invasive cercaria [20].
the complement pathway was blocked earlier than attack What are the functions of these egg secretions? The precise
complex formation at the level of C3b convertase. One further way in which the egg traverses host tissues remains a conun-
protein could play a role in immune evasion at the surface, drum. Protease activity of MW 14 and 31 kDa was reported on
namely the schistosome homologue (Smp_194920) of the host casein protease substrate gels of fractionated, unreduced eggs
T cell immunomodulatory protein. In the host it has secretions [57] but no known proteases have been linked to
Author's personal copy

R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450 1447

these activities by later proteomic analysis [59]. Omega-1 has immune polarisation via Th2 or T-regulatory pathways is the
ribonuclease activity implying that it could by cytotoxic, but demonstration, in animal models at least, of the prevention of
which host cells are the target? Egg excretion in immuno- autoimmunity (reviewed by [69]). We must presume that this
compromised mice is inhibited and can be restored by protection is a bystander effect rather than of benefit to the
immune serum or the adoptive transfer of lymphoid cells [63]. schistosome.
A proportion of eggs laid in the gut tissues may become the
centre of a granulomatous response, but the majority reach the 7. Micro-exon encoded proteins
gut lumen. Furthermore, when eggs mature, they attract leu-
cocytes to their vicinity (Fig. 2). Add to this the fact that egg The final element in this appraisal of schistosome virulence
glycans are highly immunogenic [21,22] but may serve as factors is the recent discovery of micro-exon-encoded secre-
a smoke screen rather than mediating protective responses tory proteins, collectively referred to as MEGs [29,70].
[2,23]. Thus, we can speculate that the eggs actively recruit the Currently, >70 MEGs are known, classified into 18 distinct
host immune response via their secretions, to aid their passage families with 1e23 members. They appear unique to schis-
through the tissues. Escape of the mature egg from the blood tosomes in that approximately 80% of the protein coding
vessel is the first step (see MEG section below). Does the sequence is made up of small exons ranging from 6 to 36 bp,
extensive glycosylation of IPSE and Omega-1, the major the commonest frequency being 21 bp. Microarray experi-
egg secretions, made them sticky to ensure their retention in ments have shown that the majority of MEGs are up-regulated
the eggs vicinity? Are they the agents that attract the leuco- in life cycle stages associated with establishment in the
cytic focus, potentially acting via Toll-like, C-type lectin or mammalian host after skin penetration [8]. Furthermore, RT-
mannose cell surface receptors? Host matrix metal- PCR of transcripts has revealed numerous variants generated
loproteinases, a feature of macrophage-mediated inflammation by exon skipping and, as we have seen for both larval and egg
capable of degrading all kinds of extracellular matrix proteins, secretions, these are translated to provide a pool of variant
could then provide the hydrolytic capacity. This, coupled with proteins. Unfortunately none of the MEG proteins has any
the motive power provided by smooth muscle peristalsis, homology to proteins with known functions in other organ-
would facilitate egg transit across the gut tissues. isms, and the only motif encoded is a signal sequence. Lastly,
A useful corollary to this biological process that may be WISH and immunochemistry have been used to localise
beneficial to the schistosome, is the now well-attested capacity a small number of MEGs to specific structures: MEG-3.2,
of the schistosome egg to drive T helper cell polarisation in already mentioned; MEG-4.1 to the oesophageal gland [71],
a Th2 direction [64]. Indeed, two groups have reported that and MEG-14 and MEG-4.2 to the primordia of the developing
Omega-1 is the factor responsible for polarising the immune gut in the Day 3 schistosomulum [29]. The puzzle is what
response in that direction [65,66]. However, if we are evalu- advantage the parasite gets from breaking down its coding
ating outcomes that benefit the schistosome, then there is regions into small modules, generating a range of transcripts
a contradictory element because of the claims that protective by exon skipping and then translating them into variant
immunity to schistosomes in humans is correlated with Th2 proteins? There must be an energy cost to this as not all of the
responses [67,68]. An equally intriguing consequence of variants can be functional. Is it worthwhile because it assists

Fig. 2. Multiphoton microscopy of schistosome eggs (yellow auto-fluorescence) located in venules stained with FITC-labelled Lysopersicon esculentum lectin
(green). VaDS Red B6 mice, which have CD2, CD3 red T cells were infected with schistosome cercariae 5e7 weeks earlier. The serosal collagen layer appears
blue. A, An immature egg in a venule; the red T cells are randomly distributed throughout the sub-mucosa. B, A mature egg, still within a venule, but surrounded
by an aggregate of T cells. This is a prelude to egg entry into the mucosal tissues and exit to the gut lumen. (For interpretation of the references to colour in this
figure legend, the reader is referred to the web version of this article.)
Author's personal copy

1448 R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450

Table 1

immune evasion, and if so how is that achieved? Until specific 8. Conclusions


functions have been established for one or more MEG
proteins this will be hard to authenticate. The MEG-3 family Schistosomes do not replicate in the mammalian host and
is a case in point. The proteins are secreted by the migrating lack the genetic plasticity of microbes, so thus far purposeful
skin and intravascular schistosomulum, and the mature egg products that make a schistosome infection more virulent have
also located in a blood vessel. The obvious inference is that not been defined. This means that the term virulence factor
the MEG-3 proteins interact with circulating cells, or the cannot have precisely the same meaning as for microbes.
vascular endothelium, most likely the latter. In the case of the Nevertheless, it is clear that schistosomes deliberately expose
larva, secretion from the head may first facilitate entry into proteins and glycoproteins on their surface or release them
a venule, then from the tegument, its travel through the into their host environment. Ironically the constituents of
vascular beds of internal organs to reach the portal vein. For moribund or dead eggs are the major contributors to the
the egg, secretion of MEG-3 proteins may be associated with pathogenicity of schistosomiasis, but it is hard to see how they
escape from the portal venule where it was laid, into the benefit the parasite since their release is incidental to trans-
tissues e the reverse of what the larva achieves, but still mission; hence their omission from this review. That still
involving the breach of a vessel wall. The longest isoform of leaves a significant list of candidate virulence factors
MEG-3.2 possesses 16 cysteine residues that can be aligned in (Table 2). A major task is to identify the function of such
two groups of four pairs, each separated by three amino acids candidates at the parasiteehost interface, given that significant
(Table 1). amounts of native protein are difficult or impossible to obtain
It is thus tempting to conclude that the mature protein is from the parasites themselves. RNA interference, (reviewed by
a dimer capable of cross-linking surface receptors on the [72]) is one avenue of investigation. Expression of proteins
vascular endothelium to achieve a biological effect. This like IPSE and their testing for biological effects in vitro or
should be an eminently testable proposition, provided a small in vivo is another way forward (e.g., [60]). The secreted
protein with 16 cysteines can be produced in a correctly folded proteins with multiple cysteine bonds such as those encoded
and functional form, not a trivial task. by most MEGs, may need expression in a eukaryotic system to
achieve correct folding, before testing for function. With skill,
good fortune and access to funds such studies could yield
a major dividend for our understanding of schistosomeehost
Table 2
Selected potential virulence factors. interactions.
Genome v4/v5 Genome v4/v5
Cercarial secretions Membranocalyx proteins Acknowledgements
Elastasea Smp_119130 Sm200 Smp_017730
Elastase Smp_112090 Sm29 Smp_072190 Thanks are due to Dr Adrian Mountford for commenting on
Invadolysinb Smp_090100 Sm13 Smp_195190 the manuscript, to Dr Sophie Parker-Manuel and Mr Henrique
VAL4 Smp_002070 LMWP Smp_194860
Roffato for use of their WISH images of MEG-3.2 expression
VAL18 Smp_001890 Egg secretions
VAL10 Smp_002060 IPSE Smp_112110 in the lung schistosomulum and mature egg, to Miss Hayley
Sm16 Smp_113760 Omega-1c Smp_193860 Tyrer and Mrs Jo Marrison for the multiphoton microscope
Schistosomula secretions MEG-2.1 Smp_181510 images of eggs in the intestinal wall, and Dr Mark Coles for
MEG-3.1 Smp_138080 MEG-2.2 Smp_159810 the gift of VaDS Red/B6 mice.
MEG-3.2 Smp_138070 MEG-2.5 Smp_180320
Exposed tegument enzymes MEG-2.6 Smp_180310
ATP-diphosphohydrolase 1 Smp_042020 MEG-2.8 Smp_180340 References
Phosphodiesterase Smp_153390 MEG-3.1 Smp_138080
SmNPP-5
[1] R. Bhardwaj, G. Krautz-Peterson, A. Dadara, S. Tzipori, P.J. Skelly,
Alkaline phosphatase Smp_155890 MEG-3.2 Smp_138070
Tegumental phosphodiesterase SmNPP-5 is a virulence factor for
ADP-ribosyl cyclase Smp_025830 MEG-3.3 Smp_138060
schistosomes, Infect. Immun. 79 (2011) 4276e4284.
a
Seven proteins are annotated as elastases in the latest version of the S. [2] R.A. Wilson, Proteomics at the schistosome-mammalian host interface:
mansoni genome. any prospects for diagnostics or vaccines? Parasitology 139 (2012)
b
Five genes encoding invadolysins are highly-upregulated in the intra- 1178e1194.
molluscan germ balls. [3] A. Dessein, J.C. Samuelson, A.E. Butterworth, M. Hogan, B.A. Sherry,
c
Four proteins with high homology are annotated as ribonuclease T2 M.A. Vadas, J.R. David, Immune evasion by Schistosoma mansoni: loss
(Omega-1). of susceptibility to antibody or complement-dependent eosinophil attack
Author's personal copy

R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450 1449

by schistosomula cultured in medium free of macromolecules, Parasi- [21] M. Eberl, J.A. Langermans, R.A. Vervenne, A.K. Nyame,
tology 82 (1981) 357e374. R.D. Cummings, A.W. Thomas, P.S. Coulson, R.A. Wilson, Antibodies
[4] M.A. Stirewalt, F.J. Kruidenier, Activity of the acetabular secretory to glycans dominate the host response to schistosome larvae and eggs: is
apparatus of cercariae of Schistosoma mansoni under experimental their role protective or subversive? J. Infect. Dis. 183 (2001) 1238e1247.
conditions, Exp. Parasitol. 11 (1961) 191e211. [22] T.M. Kariuki, I.O. Farah, R.A. Wilson, P.S. Coulson, Antibodies elicited
[5] G. Gazzinelli, J. Pellegrino, Elastolytic activity of Schistosoma mansoni by the secretions from schistosome cercariae and eggs are predominantly
cercarial extract, J. Parasitol. 50 (1964) 591e592. against glycan epitopes, Parasite Immunol. 30 (2008) 554e562.
[6] R.S. Curwen, R.A. Wilson, Invasion of skin by schistosome cercariae: [23] A. Van Diepen, N.S.J. Van Der Velden, C.H. Smit, M.H.J. Meevissen,
some neglected facts, Trends Parasitol. 19 (2003) 63e66. discussion C.H. Hokke, Parasite glycans and antibody-mediated immune responses
66e68. in Schistosoma infection, Parasitology 139 (2012) 1219e1230.
[7] J.P. Salter, Y. Choe, H. Albrecht, C. Franklin, K.C. Lim, C.S. Craik, [24] R.A. Paveley, S.A. Aynsley, P.C. Cook, J.D. Turner, A.P. Mountford,
J.H. McKerrow, Cercarial elastase is encoded by a functionally conserved Fluorescent imaging of antigen released by a skin-invading helminth
gene family across multiple species of schistosomes, J. Biol. Chem. 277 reveals differential uptake and activation profiles by antigen presenting
(2002) 24618e24624. cells, PLoS Negl. Trop. Dis. 3 (2009) e528.
[8] S.J. Parker-Manuel, A.C. Ivens, G.P. Dillon, R.A. Wilson, Gene [25] R.A. Paveley, S.A. Aynsley, J.D. Turner, C.D. Bourke, S.J. Jenkins,
expression patterns in larval Schistosoma mansoni associated with P.C. Cook, L. Martinez-Pomares, A.P. Mountford, The Mannose
infection of the mammalian host, PLoS Negl. Trop. Dis. 5 (2011) e1274. Receptor (CD206) is an important pattern recognition receptor (PRR) in
[9] R.S. Curwen, P.D. Ashton, S. Sundaralingam, R.A. Wilson, Identification the detection of the infective stage of the helminth Schistosoma mansoni
of novel proteases and immunomodulators in the secretions of schisto- and modulates IFN gamma production, Int. J. Parasitol. 41 (2011)
some cercariae that facilitate host entry, Mol. Cell. Proteomics 5 (2006) 1335e1345.
835e844. [26] P.C. Cook, S.A. Aynsley, J.D. Turner, G.R. Jenkins, N. Van Rooijen,
[10] A.V. Protasio, I.J. Tsai, A. Babbage, S. Nichol, M. Hunt, M.A. Aslett, N. M. Leeto, F. Brombacher, A.P. Mountford, Multiple helminth infection of
De Silva, G.S. Velarde, T.J. Anderson, R.C. Clark, C. Davidson, the skin causes lymphocyte hypo-responsiveness mediated by Th2
G.P. Dillon, N.E. Holroyd, P.T. LoVerde, C. Lloyd, J. McQuillan, conditioning of dermal myeloid cells, PLoS Pathog. 7 (2011) e1001323.
G. Oliveira, T.D. Otto, S.J. Parker-Manuel, M.A. Quail, R.A. Wilson, [27] J.E. Crabtree, R.A. Wilson, Schistosoma mansoni: an ultrastructural
A. Zerlotini, D.W. Dunne, M. Berriman, A systematically improved high examination of skin migration in the hamster cheek pouch, Parasitology
quality genome and transcriptome of the human blood fluke Schistosoma 91 (1985) 111e120.
mansoni, PLoS Negl. Trop. Dis. 6 (2012) e1455. [28] R. Harrop, R.A. Wilson, Protein synthesis and release by cultured
[11] J. Dvorak, S.T. Mashiyama, S. Braschi, M. Sajid, G.M. Knudsen, schistosomula of Schistosoma mansoni, Parasitology 107 (1993)
E. Hansell, K.C. Lim, I. Hsieh, M. Bahgat, B. Mackenzie, 265e274.
K.F. Medzihradszky, P.C. Babbitt, C.R. Caffrey, J.H. McKerrow, [29] R. DeMarco, W. Mathieson, S.J. Manuel, G.P. Dillon, R.S. Curwen,
Differential use of protease families for invasion by schistosome P.D. Ashton, A.C. Ivens, M. Berriman, S. Verjovski-Almeida,
cercariae, Biochimie 90 (2008) 345e358. R.A. Wilson, Protein variation in blood-dwelling schistosome worms
[12] J. Ingram, G. Knudsen, K.C. Lim, E. Hansell, J. Sakanari, J. McKerrow, generated by differential splicing of micro-exon gene transcripts,
Proteomic analysis of human skin treated with larval schistosome Genome Res. 20 (2010) 1112e1121.
peptidases reveals distinct invasion strategies among species of blood [30] D.J. Hockley, D.J. McLaren, Schistosoma mansoni: changes in the outer
flukes, PLoS Negl. Trop. Dis. 5 (2011) e1337. membrane of the tegument during development from cercaria to adult
[13] A. Aslam, P. Quinn, R.S. McIntosh, J. Shi, A. Ghumra, J.H. McKerrow, worm, Int. J. Parasitol. 3 (1973) 13e25.
K.A. Bunting, D.W. Dunne, M.J. Doenhoff, S.L. Morrison, K. Zhang, [31] W. Castro-Borges, A. Dowle, R.S. Curwen, J. Thomas-Oates,
R.J. Pleass, Proteases from Schistosoma mansoni cercariae cleave IgE at R.A. Wilson, Enzymatic shaving of the tegument surface of live schis-
solvent exposed interdomain regions, Mol. Immunol. 45 (2008) tosomes for proteomic analysis: a rational approach to select vaccine
567e574. candidates, PLoS Negl. Trop. Dis. 5 (2011) e993.
[14] M. Marikovsky, R. Arnon, Z. Fishelson, Proteases secreted by trans- [32] R.A. Wilson, P.S. Coulson, Immune effector mechanisms against schis-
forming schistosomula of Schistosoma mansoni promote resistance to tosomiasis: looking for a chink in the parasites armour, Trends Parasitol.
killing by complement, J. Immunol. 141 (1988) 273e278. 25 (2009) 423e431.
[15] J.P. Hewitson, Y. Harcus, J. Murray, M. van Agtmaal, K.J. Filbey, [33] J.D. Lawrence, The ingestion of red blood cells by Schistosoma mansoni,
J.R. Grainger, S. Bridgett, M.L. Blaxter, P.D. Ashton, D.A. Ashford, J. Parasitol. 59 (1973) 60e63.
R.S. Curwen, R.A. Wilson, A.A. Dowle, R.M. Maizels, Proteomic [34] S.L. Hall, S. Braschi, M. Truscott, W. Mathieson, I.M. Cesari,
analysis of secretory products from the model gastrointestinal nematode R.A. Wilson, Insights into blood feeding by schistosomes from a pro-
Heligmosomoides polygyrus reveals dominance of venom allergen-like teomic analysis of worm vomitus, Mol. Biochem. Parasitol. 179 (2011)
(VAL) proteins, J. Proteomics 74 (2011) 1573e1594. 18e29.
[16] K. Ramaswamy, B. Salafsky, S. Potluri, Y.X. He, J.W. Li, T. Shibuya, [35] Y. Jiang, X. Xue, X. Chen, W. Zhuang, J. Sun, L. Shen, W. Pan, Hae-
Secretion of an anti-inflammatory, immunomodulatory factor by Schis- mozoin from Schistosoma japonicum does not affect murine myeloid
tosomulae of Schistosoma mansoni, J. Inflamm. 46 (1995) 13e22. dendritic cell function, Parasitol. Res. 106 (2010) 653e659.
[17] P. Holmfeldt, K. Brannstrom, M.E. Sellin, B. Segerman, S.R. Carlsson, [36] T.E. Nash, A.M. Deelder, Comparison of four schistosome excretory-
M. Gullberg, The Schistosoma mansoni protein Sm16/SmSLP/SmSPO-1 secretory antigens: phenol sulfuric test active peak, cathodic circulating
is a membrane-binding protein that lacks the proposed microtubule- antigen, gut-associated proteoglycan, and circulating anodic antigen,
regulatory activity, Mol. Biochem. Parasitol. 156 (2007) 225e234. Am. J. Trop. Med. Hyg. 34 (1985) 236e241.
[18] S.J. Manuel, Patterns of Gene Expression in Schistosoma mansoni [37] A.M. Deelder, Z.L. Qian, P.G. Kremsner, L. Acosta, A.L. Rabello,
Associated with Infection of the Mammalian Host. PhD thesis, Univer- P. Enyong, P.P. Simarro, E.C. van Etten, F.W. Krijger, J.P. Rotmans, et al.,
sity of York, 2010 available from http://etheses.whiterose.ac.uk/1603/. Quantitative diagnosis of Schistosoma infections by measurement of
[19] C.H. Hokke, A.M. Deelder, K.F. Hoffmann, M. Wuhrer, Glycomics- circulating antigens in serum and urine, Trop. Geogr. Med. 46 (1994)
driven discoveries in schistosome research, Exp. Parasitol. 117 (2007) 233e238.
275e283. [38] M. Delcroix, K. Medzihradsky, C.R. Caffrey, R.D. Fetter,
[20] J. Jang-Lee, R.S. Curwen, P.D. Ashton, B. Tissot, W. Mathieson, J.H. McKerrow, Proteomic analysis of adult S. mansoni gut contents,
M. Panico, A. Dell, R.A. Wilson, S.M. Haslam, Glycomics analysis of Mol. Biochem. Parasitol. 154 (2007) 95e97.
Schistosoma mansoni egg and cercarial secretions, Mol. Cell. Proteomics [39] I.N. Crispe, The liver as a lymphoid organ, Annu. Rev. Immunol. 27
6 (2007) 1485e1499. (2009) 147e163.
Author's personal copy

1450 R.A. Wilson / Microbes and Infection 14 (2012) 1442e1450

[40] A.W. Thomson, P.A. Knolle, Antigen-presenting cell function in the Schistosoma mansoni egg secretions, Mol. Biochem. Parasitol. 155
tolerogenic liver environment, Nat. Rev. Immunol. 10 (2010) 753e766. (2007) 84e93.
[41] R.A. Wilson, P.E. Barnes, The tegument of Schistosoma mansoni: [59] W. Mathieson, R.A. Wilson, A comparative proteomic study of the
observations on the formation, structure and composition of cytoplasmic undeveloped and developed Schistosoma mansoni egg and its contents:
inclusions in relation to tegument function, Parasitology 68 (1974) the miracidium, hatch fluid and secretions, Int. J. Parasitol. 40 (2010)
239e258. 617e628.
[42] R.A. Wilson, P.E. Barnes, The formation and turnover of the mem- [60] G. Schramm, F.H. Falcone, A. Gronow, K. Haisch, U. Mamat,
branocalyx on the tegument of Schistosoma mansoni, Parasitology 74 M.J. Doenhoff, G. Oliveira, J. Galle, C.A. Dahinden, H. Haas, Molecular
(1977) 61e71. characterization of an interleukin-4-inducing factor from Schistosoma
[43] J.R. Kusel, B.H. Al-Adhami, M.J. Doenhoff, The schistosome in the mansoni eggs, J. Biol. Chem. 278 (2003) 18384e18392.
mammalian host: understanding the mechanisms of adaptation, Parasi- [61] C.M. Fitzsimmons, G. Schramm, F.M. Jones, I.W. Chalmers,
tology 134 (2007) 1477e1526. K.F. Hoffmann, C.G. Grevelding, M. Wuhrer, C.H. Hokke, H. Haas,
[44] P.J. Skelly, R. Alan Wilson, Making sense of the schistosome surface, M.J. Doenhoff, D.W. Dunne, Molecular characterization of omega-1:
Adv. Parasitol. 63 (2006) 185e284. a hepatotoxic ribonuclease from Schistosoma mansoni eggs, Mol. Bio-
[45] J.J. Van Hellemond, K. Retra, J.F. Brouwers, B.W. van Balkom, chem. Parasitol. 144 (2005) 123e127.
M. Yazdanbakhsh, C.B. Shoemaker, A.G. Tielens, Functions of the [62] G. Schramm, J.V. Hamilton, C.I. Balog, M. Wuhrer, A. Gronow,
tegument of schistosomes: clues from the proteome and lipidome, Int. J. S. Beckmann, V. Wippersteg, C.G. Grevelding, T. Goldmann, E. Weber,
Parasitol. 36 (2006) 691e699. N.W. Brattig, A.M. Deelder, D.W. Dunne, C.H. Hokke, H. Haas,
[46] S. Braschi, R.A. Wilson, Proteins exposed at the adult schistosome M.J. Doenhoff, Molecular characterisation of kappa-5, a major antigenic
surface revealed by biotinylation, Mol. Cell. Proteomics 5 (2006) glycoprotein from Schistosoma mansoni eggs, Mol. Biochem. Parasitol.
347e356. 166 (2009) 4e14.
[47] J. Mulvenna, L. Moertel, M.K. Jones, S. Nawaratna, E.M. Lovas, [63] M.J. Doenhoff, O. Hassounah, H. Murare, J. Bain, S. Lucas, The schis-
G.N. Gobert, M. Colgrave, A. Jones, A. Loukas, D.P. McManus, Exposed tosome egg granuloma: immunopathology in the cause of host protection
proteins of the Schistosoma japonicum tegument, Int. J. Parasitol. 40 or parasite survival? Trans. R. Soc. Trop. Med. Hyg. 80 (1986) 503e514.
(2010) 543e554. [64] G. Schramm, H. Haas, Th2 immune response against Schistosoma
[48] R. Bhardwaj, P.J. Skelly, Purinergic signaling and immune modulation at mansoni infection, Microbes Infect. 12 (2010) 881e888.
the schistosome surface? Trends Parasitol. 25 (2009) 256e260. [65] B. Everts, G. Perona-Wright, H.H. Smits, C.H. Hokke, A.J. van der Ham,
[49] E.G. Vasconcelos, P.S. Nascimento, M.N. Meirelles, S. Verjovski- C.M. Fitzsimmons, M.J. Doenhoff, J. van der Bosch, K. Mohrs, H. Haas,
Almeida, S.T. Ferreira, Characterization and localization of an ATP- M. Mohrs, M. Yazdanbakhsh, G. Schramm, Omega-1, a glycoprotein
diphosphohydrolase on the external surface of the tegument of Schisto- secreted by Schistosoma mansoni eggs, drives Th2 responses, J. Exp.
soma mansoni, Mol. Biochem. Parasitol. 58 (1993) 205e214. Med. 206 (2009) 1673e1680.
[50] S.P. Goodrich, H. Muller-Steffner, A. Osman, M.J. Moutin, K. Kusser, [66] S. Steinfelder, J.F. Andersen, J.L. Cannons, C.G. Feng, M. Joshi,
A. Roberts, D.L. Woodland, T.D. Randall, E. Kellenberger, P.T. LoVerde, D. Dwyer, P. Caspar, P.L. Schwartzberg, A. Sher, D. Jankovic, The major
F. Schuber, F.E. Lund, Production of calcium-mobilizing metabolites by component in schistosome eggs responsible for conditioning dendritic
a novel member of the ADP-ribosyl cyclase family expressed in Schis- cells for Th2 polarization is a T2 ribonuclease (omega-1), J. Exp. Med.
tosoma mansoni, Biochemistry 44 (2005) 11082e11097. 206 (2009) 1681e1690.
[51] R. Bhardwaj, P.J. Skelly, Characterization of schistosome tegumental [67] P. Hagan, U.J. Blumenthal, D. Dunn, A.J. Simpson, H.A. Wilkins,
alkaline phosphatase (SmAP), PLoS Negl. Trop. Dis. 5 (2011) e1011. Human IgE, IgG4 and resistance to reinfection with Schistosoma hae-
[52] H. Sprong, M. Suchanek, S.M. van Dijk, A. van Remoortere, matobium, Nature 349 (1991) 243e245.
J. Klumperman, D. Avram, J. van der Linden, J.H. Leusen, J.J. van [68] M. Webster, A.J. Fulford, G. Braun, J.H. Ouma, H.C. Kariuki,
Hellemond, C. Thiele, Aberrant receptor-mediated endocytosis of J.C. Havercroft, K. Gachuhi, R.F. Sturrock, A.E. Butterworth,
Schistosoma mansoni glycoproteins on host lipoproteins, PLoS Med. 3 D.W. Dunne, Human immunoglobulin E responses to a recombinant
(2006) e253. 22.6-kilodalton antigen from Schistosoma mansoni adult worms are
[53] S. Braschi, W.C. Borges, R.A. Wilson, Proteomic analysis of the schis- associated with low intensities of reinfection after treatment, Infect.
tosome tegument and its surface membranes, Mem. Inst. Oswaldo Cruz Immun. 64 (1996) 4042e4046.
101 (Suppl. 1) (2006) 205e212. [69] P. Zaccone, A. Cooke, Infectious triggers protect from autoimmunity,
[54] S. Braschi, R.S. Curwen, P.D. Ashton, S. Verjovski-Almeida, A. Wilson, Semin. Immunol. 23 (2011) 122e129.
The tegument surface membranes of the human blood parasite Schisto- [70] M. Berriman, B.J. Haas, P.T. LoVerde, R.A. Wilson, G.P. Dillon,
soma mansoni: a proteomic analysis after differential extraction, Pro- G.C. Cerqueira, S.T. Mashiyama, B. Al-Lazikani, L.F. Andrade,
teomics 6 (2006) 1471e1482. P.D. Ashton, M.A. Aslett, D.C. Bartholomeu, G. Blandin, C.R. Caffrey,
[55] W. Castro-Borges, D.M. Simpson, A. Dowle, R.S. Curwen, J. Thomas- A. Coghlan, R. Coulson, T.A. Day, A. Delcher, R. DeMarco, A. Djikeng,
Oates, R.J. Beynon, R.A. Wilson, Abundance of tegument surface T. Eyre, J.A. Gamble, E. Ghedin, Y. Gu, C. Hertz-Fowler, H. Hirai,
proteins in the human blood fluke Schistosoma mansoni determined by Y. Hirai, R. Houston, A. Ivens, D.A. Johnston, D. Lacerda, C.D. Macedo,
QconCAT proteomics, J. Proteomics 74 (2011) 1519e1533. P. McVeigh, Z. Ning, G. Oliveira, J.P. Overington, J. Parkhill, M. Pertea,
[56] M. Fiscella, J.W. Perry, B. Teng, M. Bloom, C. Zhang, K. Leung, R.J. Pierce, A.V. Protasio, M.A. Quail, M.A. Rajandream, J. Rogers,
L. Pukac, K. Florence, A. Concepcion, B. Liu, Y. Meng, C. Chen, M. Sajid, S.L. Salzberg, M. Stanke, A.R. Tivey, O. White, D.L. Williams,
E.C. Elgin, P. Kanakaraj, T.E. Kaufmann, J. Porter, R. Cibotti, Y. Mei, J. Wortman, W. Wu, M. Zamanian, A. Zerlotini, C.M. Fraser-Liggett,
J. Zhou, G. Chen, V. Roschke, G. Komatsoulis, B. Mansfield, S. Ruben, B.G. Barrell, N.M. El-Sayed, The genome of the blood fluke Schistosoma
I. Sanyal, T.S. Migone, TIP, a T-cell factor identified using high- mansoni, Nature 460 (2009) 352e358.
throughput screening increases survival in a graft-versus-host disease [71] G.P. Dillon, J.C. Illes, H.V. Isaacs, R.A. Wilson, Patterns of gene
model, Nat. Biotechnol. 21 (2003) 302e307. expression in schistosomes: localization by whole mount in situ
[57] P.D. Ashton, R. Harrop, B. Shah, R.A. Wilson, The schistosome egg: hybridization, Parasitology 134 (2007) 1589e1597.
development and secretions, Parasitology 122 (2001) 329e338. [72] G. Krautz-Peterson, R. Bhardwaj, Z. Faghiri, C.A. Tararam, P.J. Skelly,
[58] C.L. Cass, J.R. Johnson, L.L. Califf, T. Xu, H.J. Hernandez, RNA interference in schistosomes: machinery and methodology, Para-
M.J. Stadecker, J.R. Yates 3rd, D.L. Williams, Proteomic analysis of sitology 137 (2010) 485e495.

Vous aimerez peut-être aussi