You are on page 1of 10

Helminth Infections Decrease Host

Susceptibility to Immune-Mediated Diseases


Joel V. Weinstock and David E. Elliott
This information is current as J Immunol 2014; 193:3239-3247; ;
of April 4, 2020. doi: 10.4049/jimmunol.1400927
http://www.jimmunol.org/content/193/7/3239

References This article cites 105 articles, 33 of which you can access for free at:

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


http://www.jimmunol.org/content/193/7/3239.full#ref-list-1

Why The JI? Submit online.


• Rapid Reviews! 30 days* from submission to initial decision
• No Triage! Every submission reviewed by practicing scientists
• Fast Publication! 4 weeks from acceptance to publication
*average

Subscription Information about subscribing to The Journal of Immunology is online at:


http://jimmunol.org/subscription
Permissions Submit copyright permission requests at:
http://www.aai.org/About/Publications/JI/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
1451 Rockville Pike, Suite 650, Rockville, MD 20852
Copyright © 2014 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of
Brief Reviews Immunology
Helminth Infections Decrease Host Susceptibility to
Immune-Mediated Diseases
Joel V. Weinstock* and David E. Elliott†
Helminthic infection has become rare in highly Studies, mostly in animal models of human disease, are
industrialized nations. Concurrent with the decline in providing insight into how helminths mediate protection
helminthic infection has been an increase in the preva- from these conditions.
lence of inflammatory disease. Removal of helminths
from our environment and their powerful effects on Inflammatory bowel disease
host immunity may have contributed to this increase. Inflammatory bowel disease (IBD) is the collective term for
Several helminth species can abrogate disease in murine ulcerative colitis and Crohn’s disease. These diseases probably
models of inflammatory bowel disease, type 1 diabetes, are the consequence of an inappropriately aggressive mucosal
multiple sclerosis, and other conditions. Helminths adaptive immune response to substances normally in the in-

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


evoke immune regulatory pathways often involving testinal lumen. IBD became a significant health problem in
dendritic cells, regulatory T cells, and macrophages that highly developed countries in the 20th century, and it is
help to control disease. Cytokines, such as IL-4, IL-10, spreading in underdeveloped countries (1, 2). Epidemiologic
and TGF-b, have a role. Notable is the helminthic studies and clinic trials suggest that natural helminthic in-
modulatory effect on innate immunity, which impedes fection protects people from IBD (3).
development of aberrant adaptive immunity. Investi- Mechanisms of regulation
gators are identifying key helminth-derived immune
modulatory molecules that may have therapeutic use- Helminths modulate intestinal inflammation through activa-
fulness in the control of inflammatory disease. The tion of interactive immune regulatory circuits involving reg-
Journal of Immunology, 2014, 193: 3239–3247. ulatory T cells (Tregs), dendritic cells (DCs), macrophages,
and several cytokines (Fig. 1). There are many helminth
species inhabiting different regions of their host. Some have

H
elminths are worm-like animal parasites that have complex life cycles, traveling through the bloodstream and/or
adapted over many millions of years to live in the various tissues of the body, whereas others enter through the
gastrointestinal tract, blood, lungs, or other tissues of mouth and stay in the lumen of the gastrointestinal tract.
various species. Their long-term survival requires intricate Many only inhabit a very limited range of hosts. One may
regulatory interactions between parasite and host immunity. In expect, with wide diversity among species, that these organ-
developed countries, the 20th century brought unprecedented isms developed distinctively creative ways to modulate host
advancements in living standards associated with substantial immunity. Thus, it is surprising that evolution has endowed
improvements in agricultural practices and water and food a number of them with similar approaches to quell host im-
quality. This disrupted the life cycle of various helminths, munity.
leading to deworming of the population. The long-standing Tregs and cytokines. Animal models of IBD suggest that Tregs
close association between these parasites and their specific help to prevent excessive intestinal inflammation (4). The
hosts may have led to immune interdependency through the murine gut harbors large numbers of Foxp3+CD4+ Tregs.
process of coevolution. Epidemiologic data and animal ex- The colon and terminal ileum contain most of the intestinal
perimentation suggest that elimination of helminths con- flora. In the distal bowel, ∼25% of the lamina propria CD4+
tributes to the increasing prevalence of some immune- T cells express Foxp3, and the Foxp3+ T cells are the major
mediated diseases in regions with ever-improving sanitation. source of IL-10 (5). These cells likely function to restrain the
Diseases with increasing frequency include ulcerative colitis, host immune response to the normal intestinal flora.
Crohn’s disease, type 1 diabetes (T1D), multiple sclerosis Heligmosomoides polygyrus bakeri is a luminal murine hel-
(MS), rheumatoid arthritis (RA), asthma, and food allergy. minth that lives in the proximal small bowel, with only the

*Division of Gastroenterology, Tufts Medical Center, Boston, MA 02111; and †Division Abbreviations used in this article: AHR, airway hyperresponsiveness; CIA, collagen-
of Gastroenterology, University of Iowa, Iowa City, IA 52242 induced arthritis; CLEC, C-type lectin-like domain-containing receptor; CTLR,
C-type lectin receptor; DC, dendritic cell; DNBS, dinitrobenzene sulfonic acid; DSS,
Received for publication April 14, 2014. Accepted for publication July 7, 2014.
dextran sodium sulfate; EAE, experimental autoimmune encephalitis; IBD, inflamma-
This work was supported by National Institutes of Health Grants DK38327 and tory bowel disease; MLN, mesenteric lymph node; MS, multiple sclerosis; RA, rheuma-
DK058755, the Veterans Administration Medical Center, the Schneider family, and toid arthritis; REG, regenerating islet-derived receptor; SEA, schistosome soluble egg Ag;
the Gilman family. STZ, streptozotocin; T1D, type 1 diabetes; TNBS, trinitrobenzene sulfate; Treg, regu-
latory T cell.
Address correspondence and reprint requests to Dr. Joel V. Weinstock, Division of
Gastroenterology (Box 233), Tufts Medical Center, 800 Washington Street, Boston,
MA 02111. E-mail address: jweinstock2@tuftsmedicalcenter.org Copyright Ó 2014 by The American Association of Immunologists, Inc. 0022-1767/14/$16.00

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1400927
3240 BRIEF REVIEWS: HELMINTHS REGULATE HOST IMMUNITY

Helminthic induction of IL-10 synthesis is not an essential


factor for controlling IBD. Helminths still prevent colitis and
suppress ongoing disease in IL102/2 mice (6, 20). This
suggests that helminths activate other important immune
regulatory pathways that are IL-10/Treg independent.
Helminths also stimulate various immune cell types to
produce other cytokines that hinder the development of T cell
subtypes that are implicated in IBD pathogenesis. H. polygyrus
bakeri infection stimulates the mucosa to make TGF-b.
Transgenic mice producing T cells with disrupted TGF-bR
signaling develop colitis. In this transgenic mouse, H. poly-
gyrus bakeri infection cannot dampen the mucosal Th1 re-
sponse or prevent colitis (21). This shows that H. polygyrus
bakeri regulation of mucosal inflammation requires T cells
that respond to TGF-b.
Helminths trigger Th2-type responses that have a role in
colitis prevention. Trinitrobenzene sulfate (TNBS) mixed with
alcohol and given rectally induces murine colitis. Helminths,
FIGURE 1. Helminths activate regulatory circuits that limit inflammation
in IBD. IBD results from overresponsiveness of adaptive immune pathways to like S. mansoni and H. polygyrus bakeri, protect mice from

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


normal constituents of intestinal contents. H. polygyrus bakeri infection TNBS colitis by limiting the colonic IFN-g and IL-12 re-
induces regulatory DCs and macrophages, and it activates Tregs (CD4+ sponse. Helminths stimulate the expansion of Th2 cells that
Foxp3+) in the gut to inhibit effector T cell responses. IL-10 coming from make IL-4. Disruption of the Th2 pathway enhances Th1 cell
intestinal Tregs is particularly important. TGF-b and IL-4 also participate in differentiation and colitis, showing the importance of Th2
the regulation. Intestinal helminthic infections alter the composition of gut cytokines for disease control in this model (17). Isolated
flora. Although yet unproven, changes in intestinal flora could impact mu-
cosal immune function, leading to protection from IBD.
helminth products can stimulate these pathways. For example,
exposure to schistosome-derived recombinant glutathione S
transferase decreases TNBS-induced colitis, inhibits T cell
larval stages superficially invading the epithelial lining. This IFN-g, and promotes IL-4 and IL-10 production (M. Cap-
parasite expands the number of Foxp3+ T cells in the mes- ron, personal communication). There is a colitis model driven
enteric lymph nodes (MLNs) (6, 7) and intestinal lamina by Th2-type cytokines (oxazolone-induced colitis) in which
propria of its murine host (5). The costimulatory receptor infection with H. diminuta makes the inflammation worse
ICOS facilitates this Treg expansion (8). This helminth also (22). However, helminthic infections can curtail allergic
“activates” Tregs, making them highly regulatory (5). Rag reactions driven by the Th2 pathway (see below). Thus, there
mice reconstituted with CD252CD4+ T cells develop intes- are mechanisms of regulation independent of Th2 cytokines.
tinal inflammation as a result of a lack of Tregs. Foxp3+ IL-17 has a role in driving colitis. H. polygyrus bakeri blocks
T cells in intestines of healthy wild-type mice are not very IL-17 secretion, in part through stimulating IL-4 production
regulatory and afford no protection from colitis when trans- and, to some extent, IL-10, which affects Th17 cell function
ferred into this model of IBD. However, Foxp3+ T cells iso- (23). Disruption of Stat6 signaling specifically in T cells
lated from the colon, terminal ileum, or MLNs of H. polygyrus negates the ability of H. polygyrus bakeri infection to reverse
bakeri–infected wild-type mice populate the gut and MLNs of established CD25lo T cell transfer colitis and inhibit IL-17
the Rag recipients more readily and prevent disease (5). production (D.E. Elliott, manuscript in preparation). Expo-
H. polygyrus bakeri infection induces intestinal Tregs to sure to helminths also dampens MLN T cell responsiveness to
express several genes, as revealed using microarray and real- IL-6 through suppression of T cell IL-6Ra expression and
time PCR analysis and/or ELISA. Among these are IL-10 (5) induction of SOCS3 (D.E. Elliott, manuscript in prepara-
and GATA3 (J.V. Weinstock, unpublished observations). The tion). Induction of Th2 circuits and suppression of IL-6/Stat3
latter is noteworthy, because GATA3 is required for Tregs to signaling constrain Th17 activity.
accumulate at sites of inflammation. Moreover, it helps to CD8+ T cells also may have some role in helminthic control
sustain high-level Foxp3 and 10 expression, which are needed of IBD. After H. polygyrus bakeri infection, CD8+ Tregs can
for Tregs to protect mice from colitis (9–11). reduce the severity of colitis. They inhibit T lymphocyte
In the CD252CD4+ T cell transfer model of IBD, the proliferation through direct cell contact and class I MHC
Foxp3+IL10+CD4+ T cell subset is essential for controlling interactions without the need for IL-10 or TGF-b (24). CD8+
disease (5, 12–14). IFN-g is a driver of colitis in most IBD Tregs are implicated in the control of several diseases featur-
models. In the gut, H. polygyrus bakeri–activated Tregs con- ing immune dysregulation (25, 26).
trol colitis, in part through secretion of IL-10, which inhibits Regulatory DCs. Using another model of colitis, it was shown
the production of IFN-g from mucosal effector T cells. Other that helminths also control IBD through activation of intes-
mechanisms of action are likely as well. As with H. polygyrus tinal regulatory DCs. T cell– and B cell–deficient Rag mice
bakeri infection (15), other helminthic species, such as reconstituted with IL102/2 T cells develop colitis because
Hymenolepis diminuta (16) and Schistosoma mansoni (17), also their Tregs cannot make IL-10. Rag mice infected with
induce IL-10 secretion. Litomosoides sigmodontis suppressed H. polygyrus bakeri and then dewormed with a pharmaceutical
B cell responses in the host through induction of IL-10 and agent before IL102/2 T cell reconstitution are protected
Tregs (18, 19). from colitis (20). Moreover, the intestinal mucosa makes
The Journal of Immunology 3241

less colitogenic cytokines, such as IFN-g and IL-17, after this on or in DCs alters their function. There are four families of
brief H. polygyrus bakeri exposure, even if the animals are such receptors that include TLRs and C-type lectin receptors
configured to remain free from colitis. This infers that (CTLRs). Microarray analysis revealed that the intestinal DCs
H. polygyrus bakeri can act through cells of innate immunity expressed several TLRs, and H. polygyrus bakeri infection
to render animals resistant to disease. decreased the expression of several TLR subtypes (9, 13).
The mechanism underlying this protection involves in- Also, there is downmodulation of LPS-binding protein and
duction of regulatory DCs in the intestinal mucosa (20). CD14, important components of the TLR4-signaling com-
Activation of these cells does not require the aid of T or plex (Fig. 2).
B cells. Compared with DCs from uninfected animals, in- Two classes of CTLRs are regenerating islet-derived
testinal DCs after H. polygyrus bakeri infection only weakly receptors (REGs) and C-type lectin-like domain-containing
support Ag-driven IFN-g secretion. Furthermore, DCs iso- receptors (CLECs). REGs are secretory proteins that act on
lated from the intestines or MLNs of H. polygyrus bakeri– the structure bearing the ligand, without modulating the
infected Rag mice transferred into colitis-susceptible mice function of their cell of origin (29). Secretory REGs, like 3b
block colitis and mucosal Ag–induced IFN-g and IL-17 and 3g, bind intestinal bacteria and other organisms, leading
responses (27). to their demise (30, 31). REG2/2 mice show the importance
The mechanism through which these regulatory DCs quell of some of these REGs (30). H. polygyrus bakeri infection
colitis has been characterized in part. The regulatory DCs do greatly increases the expression of all REGS (REG1, REG3a,
not prevent effector T cells from populating the gut or MLNs, REG3b, REG3g, REG4) displayed by intestinal DCs. High
but they do inhibit their function. The regulatory DCs, REG protein secretion by intestinal DCs would help to keep

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


through a cell contact–dependent mechanism, interfere with organisms away from the DC membrane. REG4 has anti-
the interaction of proinflammatory DCs with their effector apoptosis properties that protect host cells from death (32,
T cell counterparts. This prevents Ag-induced IFN-g and IL- 33).
17 secretion. IL-4, IL-10, and TGF-b, as well as Tregs, are Host cells express CLECs as transmembrane proteins.
not essential for this regulatory process (27). When CLECs engage their ligands, the receptors trigger
H. polygyrus bakeri residing in the proximal bowel induces intracellular-signaling pathways to alter cell function (34).
substantial changes in the activation state of DCs residing in CLEC7A engages components of fungi and some bacteria,
the distal intestine (Fig. 2). Microarray analysis shows sub- whereas CLEC9A binds dead or dying cells (35). When en-
stantial downmodulation of Jak1/2 and other intracellular gaged, some CLEC receptors, like CLEC7A and CLEC9A,
signaling pathways (several MAPKs) important for the in- activate DCs, promoting T cell activation (35). H. polygyrus
duction of proinflammatory cytokines that drive effector bakeri infection profoundly inhibits the expression of nearly
T cell activation. Many components of the MHC Ag-presenting all of the CLECs expressed by intestinal DCs (4N, 7A, 9A,
complex (e.g., CD40, CD80, CD86, H2) are also reduced. and 12A). Therefore, decreased CLEC (e.g., 7A, 9A) ex-
In addition, the DCs display decreased expression for mol- pression associated with heightened REG secretion makes it
ecules associated with the receptor-signaling pathways for less likely that organisms and necrotic cells will approach DCs
IL-1, CSF, IL-6, and TGF-b (20) (J.V. Weinstock, un- and encounter membrane-bound, proinflammatory CLEC
published observations). receptors. Thus, helminthic regulation of TLRs and CTLRs
An intriguing discovery is the effect of H. polygyrus bakeri (CLECs and REGs) may render intestinal DCs less likely to
infection on intestinal DC innate immune receptor expres- activate adaptive immunity and, subsequently, IBD.
sion. DCs are the critical link between innate and adaptive Regulatory macrophages. Helminths also protect from IBD
immunity (28). They sample Ags in the intestinal lumen and through induction of alternatively activated macrophages.
present these Ags to T cells, inducing their differentiation and Helminths induce the production of IL-10 and Th2 cytokines,
proliferation or perhaps rendering them inert. DCs sense such as IL-4 and IL-5, which activate macrophages in distinct
threats in the environment through germline-encoded pattern ways (36). These alternatively activated macrophages make
recognition receptors that bind motifs on bacteria, fungi, IL-10, TGF-b, and other immunomodulatory factors that
viruses, or stressed hosts cells. Engagement of these receptors can modulate Th1-type inflammation (37).

FIGURE 2. Effects of H. polygyrus bakeri infection on


the function of DCs. H. polygyrus bakeri infection blocks
CLEC and TLR expression and promotes REG secretion
in gut DCs. The infection also inhibits DC intracellular
signaling pathways (Jak1 and Jak2, and several MAPKs)
important for proinflammatory cytokine production.
There also is disruption in the signaling pathways for IL-1,
IL-6, TGF-b, and CSF. The MHC complex is down-
modulated as well (CD40, CD80, CD86, MHCII). As
a result of these changes, intestinal DCs are less able to
activate effector T cells.
3242 BRIEF REVIEWS: HELMINTHS REGULATE HOST IMMUNITY

Another model of IBD is dextran sodium sulfate (DSS)- testinal epithelium releases regulatory molecules and sits close
induced enteritis. DSS administered orally to rodents dam- to immunocytes. Infection with Trichuris muris stimulates
ages the intestinal epithelial lining, inducing gut inflammation. intestinal epithelial cells to make thymic stromal lympho-
Infection of BALB/c mice with S. mansoni protects from poietin, which can interact with the lamina propria DCs,
DSS-induced injury through induction of regulatory macro- promoting a Th2 response and worm expulsion. It also limits
phages. The adult schistosome flukes, living in the portal vein, IL-12 and IFN-g production in DSS-induced colitis, reduc-
induce this protection. The protective process does not re- ing pathology (51).
quire Tregs or regulatory cytokines, such as TGF-b and IL-10 Although some intestinal helminths have no fixed associa-
(38). tion with the epithelial lining, others interact closely with the
A cysteine protease inhibitor (cystatin) of filarial nematodes mucosal barrier (e.g., hookworm) or place holdfasts beneath
protects mice from DSS-induced colitis (39). Macrophages the epithelial lining (e.g., whipworm). This affords further
and IL-10 are necessary for this protection, as suggested by access for direct communication with T cells to induce Tregs
a lung inflammatory model. Cystatin activates intracellular- (7) or with other cells to promote regulation (49).
signaling pathways, such as ERK and p38, which induce Gut bacteria are important for the health of the mucosal
macrophages to make IL-10 and IL-12p40 (40). immune system and readily interact with intestinal DCs and
In the IL102/2 Rag model of IBD, H. polygyrus bakeri other cells (52). H. polygyrus bakeri infection modifies the
infection induces regulatory macrophages in the gut of Rag distribution and abundance of some intestinal bacteria. There
mice. These cells are induced even if mice are not recon- is an increase in Lactobacillaceae. Various bacterial species
stituted with T or B cells. Thus, this induction does not within this group inhibit intestinal inflammation in models of

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


require participation of adaptive immunity. These intesti- colitis (53). Rhesus monkeys develop colitis. Trichuris tri-
nal macrophages inhibit Ag-induced IL-17 and IFN-g se- chiura infection results in a milder colitis associated with
cretion by a contact-dependent mechanism. Also, when reduced bacterial attachment to the epithelial surface and
transferred into Rag mice, they protect animals from colitis changes to the composition of microbial communities at-
(J.V. Weinstock, A.M. Blum, and L. Hang, unpublished tached to the intestinal mucosa (54).
observations). Helminths also may protect via enhancement of mucosal
In another study using dinitrobenzene sulfonic acid (DNBS) barrier function (55). Trichuris infection stimulates IL-22
instead of TNBS to induce IBD, infection with H. diminuta production in the mucosa, which is a molecule associated
protects mice from colitis through induction of alternatively with epithelial repair and enhancement of the overlying mu-
activated macrophages in the colon. Alternatively activated cous layer (54, 56).
macrophages transferred into mice protect from DNBS- There are helminth species that suppress colitis while living
induced injury, attesting to their role in the regulatory pro- in regions of the host distant from the intestines. Their mode of
cess. Extracts from H. diminuta worms injected i.p. also communication with host immunity could be different. For
provide protection and suppress macrophage function in vitro example, the filaria Brugia malayi resides in lymphatics and
(41, 42). releases copious amounts of asparaginyl-tRNA synthetase,
Thus, macrophages activated by helminth infection can which can block IL-8 signaling in human and murine leu-
suffice to protect from IBD (42). This also suggests that some kocytes and suppress murine T cell–transfer colitis (57).
helminths make soluble factors that can mediate this process In summary, animal models suggest that helminths control
in lieu of live organisms. In the DNBS model, alternatively colitis via induction of several distinct immune-regulatory
activated macrophages work through an IL-10–dependent pathways. This includes promotion of Treg function through
mechanism to control colitis (43). induction of Gata3, IL-33R, and IL-10 expression, generation
Communication with the host and penetrating the epithelial barrier. of regulatory DCs with a unique phenotype (Fig. 2), and in-
To modulate colitis, helminths must release soluble factors or duction of regulatory macrophages. Also, it appears that
communicate with the host in some other fashion. The IL-4 and TGF-b, as well as their signaling pathways, have
presence of worm-derived soluble factors is supported by a role. However, these regulatory pathways are not necessarily
experiments that use extracts from H. diminuta worms (41) called into play simultaneously with similar importance in each
or dead schistosome OVA to protect mice from colitis (17). distinct IBD animal model.
Helminths produce a number of products with immune-
Helminths and other immune-mediated diseases
modulatory properties (7, 44–49). For instance, helminths
produce molecules that induce Tregs (7). To induce regulatory Genome-wide association studies demonstrated susceptibility
cells, intestinal helminths must breach the mucosal barrier to gene overlap between IBD and other autoimmune and
engage the immune system. This communication may occur immune-mediated inflammatory diseases (58). MS, T1D, RA,
through several possible mechanisms. and asthma, like IBD, have emerged in populations benefiting
DCs advance dendrites across the epithelial barrier, which from advanced socioeconomic development. This suggests
could permit intestinal helminths in the intestines to com- that the environmental factors that impacted immune path-
municate directly with these cells. Supporting this hypothesis ways and increased the risk for IBD also increased the risk for
are data showing that H. polygyrus bakeri and other helminths other immune-mediated illnesses. Animal models of these
release factors that affect the state of DC activation (49, 50). organ-specific inflammatory diseases show that many of the
This, in turn, can result in decreased Ab responses (50) and helminth-induced regulatory circuits that regulate murine
stimulation of Treg development. colitis suppress inflammation in these diseases as well.
Direct interactions between intestinal helminths and the gut Animal models of MS. Mice or rats immunized with
epithelium is another possible mechanism of action. The in- myelin-associated Ags develop experimental autoimmune
The Journal of Immunology 3243

encephalitis (EAE), a model of MS (59). Mice exposed different classes of helminths evolved independently and may
to viable S. mansoni or dead OVA are protected from use different products to influence immune-regulatory path-
developing EAE (60, 61). Schistosome exposure suppresses ways. It will be interesting to determine how divergent or
splenocyte and CNS cell production of IL-12p40, IFN-g, convergent are the mechanisms used by different helminths.
and TNF-a while increasing TGF-b, IL-10, and IL-4. Animal models of T1D. T1D develops spontaneously in auto-
Infection with H. polygyrus bakeri (62) or Fasciola hepatica immune-prone NOD mice, or it can be elicited after serial
(63) or treatment with soluble Trichuris suis adult or larval injection of low-dose streptozotocin (STZ; an islet b cell
Trichinella spiralis homogenate (64) also suppresses EAE toxin) in other strains (71). Infection with S. mansoni,
disease scores, with similar changes in the cytokine profile. T. spiralis, H. polygyrus bakeri, or L. sigmodontis protects NOD
T. spiralis infection also affords protection in a rat EAE model mice from insulitis (72–75). However, the mechanisms of protec-
(65). Draining popliteal lymph node cells from parasite- tion may differ among species.
exposed rats produce less IFN-g and IL-17 and more IL-10 Young NOD mice exposed to schistosome OVA alone or to
and IL-4 in response to concanavalin A stimulation compared SEA are protected from developing diabetes. SEA-induced
with cells from helminth-naive animals. Infection also protection is associated with increased pancreatic mononu-
increases the number of CD4+CD25+Foxp3+ T cells in the clear cell expression of TGF-b, IL-4, and IL-10 mRNA (76).
spleen. Adoptive transfer of splenic T cells from infected rats SEA treatment increases the number of CD4+CD25+Foxp3+
into helminth-naive rats protects recipients from developing T cells in the pancreas and spleen. Splenocytes from SEA-
EAE (65). treated NOD mice do not produce disease when transferred
As discussed above, helminths produce factors that mediate into NOD.scid recipients (76). Depletion of Tregs from these

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


this protection. Adoptive transfer of bone marrow–derived splenocytes restores their pathogenicity, showing that this is
DCs exposed to excretory/secretory products from cultured a critical pathway for protection.
T. spiralis muscle cyst larvae also protects against EAE (66). Intraperitoneal injection of excretory/secretory Ags from
Protection is associated with decreased DC IL-12p70 pro- F. hepatica also provides protection from insulitis in NOD
duction and increased DC IL-10 production. Splenocytes mice (77). Disease prevention is associated with the induction
from rats that receive helminth product–exposed DCs prior to of IL-10–secreting B cells and transcripts indicative of M2
EAE challenge have more Foxp3+ T cells, make less IL-17A macrophage induction in pancreatic lymph node cell pop-
and IFN-g, and produce more IL-4, IL-10, and TGF-b than ulations.
do splenocytes from rats that receive medium-alone–exposed Infection with L. sigmodontis delays diabetes in IL-4–defi-
DCs (66). cient NOD mice and is associated with increased numbers of
Infection with Taenia crassiceps also inhibits development of splenic CD4+CD25+Foxp3+ Tregs. Like splenocytes from
EAE (67). Inhibition is associated with suppression of TNF-a SEA-treated NOD mice, splenocytes from L. sigmodontis–
and induction of alternatively activated macrophages. Factors infected mice do not produce diabetes when transferred into
released by T. crassiceps cysticerci impair LPS-stimulated bone NOD.scid recipients. However, unlike the SEA model, de-
marrow–derived IL-12 and TNF-a production in a cRAF- pletion of Tregs does not restore pathogenicity, suggesting
dependent manner (68). that this is not a critical pathway for L. sigmodontis–mediated
Clinical studies involving patients with MS from helminth- protection. Instead, blockade of TGF-b function abrogates
endemic areas show that those with active helminthic infec- protection, indicating that this pathway is critical for this
tions have attenuated disease compared with uninfected helminth and disease model (75).
patients. Treatment of helminthic infections results in wors- Infection with H. polygyrus bakeri also protects IL-4–defi-
ening MS activity that is associated with an increase in the cient NOD mice from developing diabetes (78). Protection is
fraction of PBMCs making IFN-g and IL-12 and a decrease in associated with induction of IL-10 production by CD127hi
the fraction producing IL-10 and TGF-b (69). Helminth Foxp32 T cells present in pancreatic lymph nodes. Blockade
removal also decreases the frequency of circulating CD4+ of IL-10 function in vivo in IL-4–deficient (but not IL-4–
CD25+Foxp3+ T cells. Patients with MS and active helminth sufficient) NOD mice abrogates protection (78). This sug-
infections had an increased frequency of spinal fluid Foxp3+ gests that helminth-amplified IL-4 and IL-10 circuits can
Treg cells and higher serum retinoic acid levels compared with independently provide protection.
healthy controls or uninfected MS patients (70). Exposure of Infection with T. crassiceps decreases insulitis and protects
PBMC-derived DCs to schistosome soluble egg Ags (SEAs) BALB/C and C57BL/6 mice from STZ-induced diabetes.
induced enzymes involved in retinoic acid synthesis, likely by T. crassiceps–induced protection is associated with an increase
a TLR2 activation–dependent pathway. LPS-stimulated DCs in IL-4 and alternatively activated macrophages but not with
derived from PBMCs of helminth-infected patients made induction of Tregs (79).
less IL-6, IL-12p70, IL-23, and TNF-a than did DCs Infection with H. polygyrus bakeri also affords protection
from uninfected patients, and levels were further reduced, from STZ-induced diabetes in C57BL/6 mice (80). H. poly-
in a SOCS3-dependent fashion, by exposure to SEA. SEA- gyrus bakeri–induced protection remains intact in STAT6- or
exposed DCs, cocultured with autologous CD252 T cells, re- IL-10–deficient mice, suggesting that IL-4 and IL-10, indi-
duce T cell STAT3 activation while increasing SMAD3 activa- vidually, are not required for protection in this model.
tion and Foxp3 expression (70). These studies show that helminths residing in different host
These studies show that helminths can suppress other organ- tissues can suppress the same organ-specific inflammation.
specific inflammatory diseases beyond colitis. In addition, they Critical mechanisms for protection from T1D appear to vary
show that infection with diverse helminths (nematodes, among various helminthic species. Also, they use mechanisms
trematodes, and cestodes) can suppress a specific disease. These that differ from those that protect from colitis or EAE. This
3244 BRIEF REVIEWS: HELMINTHS REGULATE HOST IMMUNITY

may reflect the varying importance of specific regulatory inflammation in the K/BxN model, and worsening of arthritis
pathways for each model system. is likely due to helminth-induced mast cell activation (91).
Animal models of RA. Collagen-induced arthritis (CIA) is These studies again confirm the broad nature of helminth-
a murine model of RA that develops in mice immunized associated immune regulation. In addition, they demonstrate
with type II collagen in CFA (81). Infection with S. mansoni the central protective roles for IL-10 and TGF-b production
before collagen sensitization protects mice from developing and suppression of IFN-g and IL-17 circuitry by helminths in
polyarticular arthritis (82). This protection is associated with control of arthritis.
reduced IFN-g, TNF-a, and IL-17 production, but increased Animal models of allergy/asthma. Animal models and clinical
IL-4 and IL-10 production, by splenocytes compared with studies indicate that dysregulated Th1/Th17 responses
collagen-sensitized helminth-naive mice (82). Schistosoma underlie IBD, MS, T1D, and RA. Because helminth infection
japonicum also protects mice from CIA. The infection results directly suppresses those cytokine pathways, helminth-associated
in suppressed IFN-g secretion, but augmented IL-4 and IL-10 suppression of these diseases appears to be somewhat straight-
secretion, by mitogen-stimulated splenocytes (83, 84). forward.
Another rodent arthritis model is monoarticular inflamma- In contrast, allergy and asthma appear to result from ex-
tion provoked by injection of CFA (without collagen) into a knee cessive Th2-type inflammation. Because helminth infections
joint. Treatment with a 16-kDa recombinant protein derived usually stimulate strong Th2 responses, it is counterintuitive
from S. japonicum (rSj16) protects rats from CFA-induced joint that helminthic exposure would lessen allergic inflammation.
inflammation. Protection is associated with a reduction in se- However, studies comparing groups treated to remove hel-
rum TNF-a, NO, and IL-1b and restoration of IL-10 levels minths with untreated controls suggest that helminth infection

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


compared with untreated arthritic and control rats (85). decreases the prevalence of atopy, at least as measured by skin
A 62-kDa phosphocholine-containing glycoprotein (ES-62) prick test positivity (92).
isolated from Acanthocheilonema viteae prevents and treats Animal models indicate that helminths induce regulatory
established CIA (86). Collagen-stimulated draining lymph pathways that can suppress atopic disease. A major murine
node cells from ES-62–treated mice make less IFN-g and model of allergic inflammation is airway hyperresponsiveness
TNF-a and more IL-10 than do cells from untreated mice. (AHR) induced by respiratory exposure to an Ag previously
Treatment of mice with ES-62 before CIA induction results used to sensitize the animals (93). This sensitization uses Ag
in reduced serum IL-17 levels and fewer Th17 cells in the mixed with alum adjuvant.
draining lymph node and affected joints (87). Bone marrow– Infection with H. polygyrus bakeri during or prior to OVA
derived DCs stimulated with LPS and ES-62 make less sensitization inhibits subsequent airway reactivity (94) and
TNF-a, IL-6, and IL-23 and have reduced ability to induce inflammation (94, 95) upon aerosol challenge. Transfer of
IL-17 production by OT-II T cells in vitro. ES-62 also works MLN cells or splenocytes from infected mice into helminth-
directly on polarized Th17 cells to reduce IL-17 production naive animals inhibits airway inflammation, showing activa-
and MyD88 expression (87). A synthetic small molecule tion of regulatory cells. H. polygyrus bakeri exposure increases
modeled after ES-62 (N-(2-[(4-bromobenzyl)sulfonyl]ethyl)- the percentage of CD4+CD25+Foxp3+ T cells in the mesenteric
N,N-dimethylamine, 11a) inhibits development of CIA and and thoracic lymph nodes (94, 95). In addition, H. polygyrus
suppresses the release of IL-12p40 and IL-6 from LPS- bakeri colonization induces a CD19+CD23+ regulatory B cell
stimulated macrophages (88). population that can adoptively transfer suppression of airway
Culturing CpG-stimulated bone marrow–derived DCs with inflammation, independent of IL-10 production (62). Excretory/
F. hepatica total extract increases IL-10 and TGF-b produc- secretory products from cultured adult H. polygyrus bakeri worms
tion and suppresses IL-12p70, IL-23, IL-6, and TNF-a also inhibit OVA-stimulated airway inflammation and hyperre-
production compared with DCs stimulated without extract activity when given at the time of OVA alum sensitization (96).
(89). F. hepatica extract–treated DCs pulsed with collagen and This protection is associated with reduced IL-4, IL-5, IL-13,
then given to mice suppress CIA and inhibit IL-17 and IFN-g and IFN-g levels in bronchoalveolar lavage fluid, suppression
but augment IL-4, IL-10, and TGF-b production by of the OVA-induced increase in alternatively activated macro-
collagen-stimulated draining lymph node cells. Extract- phage markers, and reduced Teffector/Treg ratios in lung
treated DCs increase the frequency of CD25+Foxp3+ Tregs tissue (96).
in draining lymph nodes, and transfer of these T cells sup- Exposure to other helminths, such as S. mansoni (97–99),
pressed CIA in recipients (89). S. japonicum (100), or T. spiralis (101), also affords protection
H. diminuta infection before intra-articular CFA challenge from allergic airway reactivity and inflammation. Helminth
reduces joint swelling and speeds the resolution of inflam- exposure is associated with decreased OVA-stimulated IL-4
mation in mice (90). H. diminuta exposure suppresses CFA- and IL-5, but increased IL-10 and TGF-b production, as
induced TNF-a mRNA expression in challenged joints. In- measured in either bronchoalveolar lavage fluid or super-
fection increases splenocyte IL-4 and IL-10 production, and natants from cultured pulmonary draining lymph node cells
H. diminuta infection does not protect IL-10–deficient mice or splenocytes. Splenic CD11c+ DCs isolated from S. japo-
from CFA arthritis (90), suggesting that IL-10 is important nicum–infected mice transfer protection to helminth-naive
for this protection. mice (100). Transfer of splenic T cells from T. spiralis–
The same group found that H. diminuta worsens joint in- infected mice, which contain .2-fold higher percentages of
flammation in another arthritis model in which disease results CD4+CD25+Foxp3+ cells, provides partial protection against
from injecting BALB/c mice with serum from arthritogenic OVA-induced airway inflammation (102). S. mansoni infec-
K/BxN mice that contains Abs against autologous glucose-6- tion also induces CD4+CD25+Foxp3+ Tregs, and targeted in
phosphate isomerase (91). Mast cells are required for joint vivo depletion of Foxp3-expressing cells negates the protective
The Journal of Immunology 3245

influence of infection, supporting the importance of Tregs for 3. Weinstock, J. V., and D. E. Elliott. 2013. Translatability of helminth therapy in
inflammatory bowel diseases. Int. J. Parasitol. 43: 245–251.
this protection (99). In addition, like H. polygyrus bakeri, 4. MacDonald, T. T., I. Monteleone, M. C. Fantini, and G. Monteleone. 2011.
exposure to S. mansoni induces CD19+CD23+ regulatory Regulation of homeostasis and inflammation in the intestine. Gastroenterology 140:
1768–1775.
B cells that can transfer protection from AHR (103). These 5. Hang, L., A. M. Blum, T. Setiawan, J. P. Urban, Jr., K. M. Stoyanoff, and
regulatory B cells express CD1d, require intact IL-10 pro- J. V. Weinstock. 2013. Heligmosomoides polygyrus bakeri infection activates colonic
duction, and act, in part, by increasing the number of pul- Foxp3+ T cells enhancing their capacity to prevent colitis. J. Immunol. 191: 1927–
1934.
monary CD4+CD25+Foxp3+ Tregs in the lungs (103). 6. Elliott, D. E., T. Setiawan, A. Metwali, A. Blum, J. F. Urban, Jr., and
Treatment of mice with ES-62 from A. viteae also protects J. V. Weinstock. 2004. Heligmosomoides polygyrus inhibits established colitis in IL-
10-deficient mice. Eur. J. Immunol. 34: 2690–2698.
from OVA-AHR and pulmonary inflammation (46, 104) in 7. Grainger, J. R., K. A. Smith, J. P. Hewitson, H. J. McSorley, Y. Harcus,
association with reduced mast cell degranulation, lower OVA- K. J. Filbey, C. A. Finney, E. J. Greenwood, D. P. Knox, M. S. Wilson, et al.
2010. Helminth secretions induce de novo T cell Foxp3 expression and regulatory
stimulated IL-4, IL-5, and IL-13 production by draining function through the TGF-b pathway. J. Exp. Med. 207: 2331–2341.
lymph node cells, and decreased Th17 cells compared with 8. Redpath, S. A., N. van der Werf, A. M. Cervera, A. S. MacDonald, D. Gray,
ES-62–naive mice. Treatment with anti–IFN-g abrogates R. M. Maizels, and M. D. Taylor. 2013. ICOS controls Foxp3(+) regulatory T-cell
expansion, maintenance and IL-10 production during helminth infection. Eur. J.
protection from airway inflammation/reactivity and reverses Immunol. 43: 705–715.
changes in cytokine profile and Th17 frequency (46), indi- 9. Wohlfert, E. A., J. R. Grainger, N. Bouladoux, J. E. Konkel, G. Oldenhove,
C. H. Ribeiro, J. A. Hall, R. Yagi, S. Naik, R. Bhairavabhotla, et al. 2011. GATA3
cating that ES-62 induces counter-regulatory Th1 circuitry in controls Foxp3+ regulatory T cell fate during inflammation in mice. J. Clin. Invest.
this model. 121: 4503–4515.
10. Wang, Y., M. A. Su, and Y. Y. Wan. 2011. An essential role of the transcription
factor GATA-3 for the function of regulatory T cells. Immunity 35: 337–348.
Conclusions 11. Shoemaker, J., M. Saraiva, and A. O’Garra. 2006. GATA-3 directly remodels the

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


Helminth infections exerted a strong selective pressure on IL-10 locus independently of IL-4 in CD4+ T cells. J. Immunol. 176: 3470–3479.
12. Schmitt, E. G., D. Haribhai, J. B. Williams, P. Aggarwal, S. Jia,
our genome (105). Many host factors that confer risk for L. M. Charbonnier, K. Yan, R. Lorier, A. Turner, J. Ziegelbauer, et al. 2012. IL-10
immune-mediated disease evolved under the selection pres- produced by induced regulatory T cells (iTregs) controls colitis and pathogenic ex-
iTregs during immunotherapy. J. Immunol. 189: 5638–5648.
sure of helminths (106). Thus, it is plausible that eradication 13. Rubtsov, Y. P., R. E. Niec, S. Josefowicz, L. Li, J. Darce, D. Mathis, C. Benoist,
of helminthic infections and the loss of their immune- and A. Y. Rudensky. 2010. Stability of the regulatory T cell lineage in vivo. Science
329: 1667–1671.
modulatory effects promoted the development of some of 14. Uhlig, H. H., J. Coombes, C. Mottet, A. Izcue, C. Thompson, A. Fanger,
the immunological diseases. A. Tannapfel, J. D. Fontenot, F. Ramsdell, and F. Powrie. 2006. Characterization
There are numerous animal models representing a diverse of Foxp3+CD4+CD25+ and IL-10-secreting CD4+CD25+ T cells during cure of
colitis. J. Immunol. 177: 5852–5860.
range of diseases for which helminths prevent and/or abrogate 15. Setiawan, T., A. Metwali, A. M. Blum, M. N. Ince, J. F. Urban, Jr., D. E. Elliott,
inflammation in various organs. Many helminth species me- and J. V. Weinstock. 2007. Heligmosomoides polygyrus promotes regulatory T-cell
cytokine production in the murine normal distal intestine. Infect. Immun. 75:
diate protection, evoking similar immune-regulatory mecha- 4655–4663.
nisms. Common themes include modulation of DC function, 16. Hunter, M. M., A. Wang, C. L. Hirota, and D. M. McKay. 2005. Neutralizing
anti-IL-10 antibody blocks the protective effect of tapeworm infection in a murine
activation of Tregs, alteration of macrophage activity, and model of chemically induced colitis. J. Immunol. 174: 7368–7375.
enhancement of regulatory cytokine synthesis. Several of these 17. Elliott, D. E., J. Li, A. Blum, A. Metwali, K. Qadir, J. F. Urban, Jr., and
mechanisms appear to function concurrently and indepen- J. V. Weinstock. 2003. Exposure to schistosome eggs protects mice from TNBS-
induced colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 284: G385–G391.
dently of each other. Thus, the loss of any one regulatory 18. Hartmann, W., I. Haben, B. Fleischer, and M. Breloer. 2011. Pathogenic nem-
pathway will not necessarily abrogate protection from disease. atodes suppress humoral responses to third-party antigens in vivo by IL-10-
mediated interference with Th cell function. J. Immunol. 187: 4088–4099.
A complex array of different gene interactions, environmental 19. Haben, I., W. Hartmann, S. Specht, A. Hoerauf, A. Roers, W. M€uller, and
factors, and aberrant host immune responses drive immuno- M. Breloer. 2013. T-cell-derived, but not B-cell-derived, IL-10 suppresses antigen-
specific T-cell responses in Litomosoides sigmodontis-infected mice. Eur. J. Immunol.
logical diseases. Thus, the mechanisms of protection should 43: 1799–1805.
not be expected to be the same for all diseases, mouse strains, 20. Hang, L., T. Setiawan, A. M. Blum, J. Urban, K. Stoyanoff, S. Arihiro,
and humans. The vast array of independent regulatory circuits H. C. Reinecker, and J. V. Weinstock. 2010. Heligmosomoides polygyrus infection
can inhibit colitis through direct interaction with innate immunity. J. Immunol.
that helminths engage may explain why they affect many 185: 3184–3189.
disease states. 21. Ince, M. N., D. E. Elliott, T. Setiawan, A. Blum, A. Metwali, Y. Wang,
J. F. Urban, Jr., and J. V. Weinstock. 2006. Heligmosomoides polygyrus induces
A number of investigations are underway to identify the TLR4 on murine mucosal T cells that produce TGFbeta after lipopolysaccharide
helminth-derived molecular signals that mediate host immune stimulation. J. Immunol. 176: 726–729.
22. Wang, A., M. Fernando, G. Leung, V. Phan, D. Smyth, and D. M. McKay. 2010.
modulation. This could lead to new pharmaceutical agents that Exacerbation of oxazolone colitis by infection with the helminth Hymenolepis
target unique immune regulatory pathways, which will allow diminuta: involvement of IL-5 and eosinophils. Am. J. Pathol. 177: 2850–2859.
safe control or the prevention of some immune-mediated 23. Elliott, D. E., A. Metwali, J. Leung, T. Setiawan, A. M. Blum, M. N. Ince,
L. E. Bazzone, M. J. Stadecker, J. F. Urban, Jr., and J. V. Weinstock. 2008.
illnesses. Colonization with Heligmosomoides polygyrus suppresses mucosal IL-17 production.
J. Immunol. 181: 2414–2419.
24. Metwali, A., T. Setiawan, A. M. Blum, J. Urban, D. E. Elliott, L. Hang, and
Disclosures J. V. Weinstock. 2006. Induction of CD8+ regulatory T cells in the intestine by
Heligmosomoides polygyrus infection. Am. J. Physiol. Gastrointest. Liver Physiol. 291:
The authors have no financial conflicts of interest. G253–G259.
25. Leavenworth, J. W., X. Tang, H. J. Kim, X. Wang, and H. Cantor. 2013.
Amelioration of arthritis through mobilization of peptide-specific CD8+ regulatory
References T cells. J. Clin. Invest. 123: 1382–1389.
1. Molodecky, N. A., I. S. Soon, D. M. Rabi, W. A. Ghali, M. Ferris, G. Chernoff, 26. Kim, H. J., and H. Cantor. 2011. Regulation of self-tolerance by Qa-1-restricted
E. I. Benchimol, R. Panaccione, S. Ghosh, H. W. Barkema, and G. G. Kaplan. CD8(+) regulatory T cells. Semin. Immunol. 23: 446–452.
2012. Increasing incidence and prevalence of the inflammatory bowel diseases with 27. Blum, A. M., L. Hang, T. Setiawan, J. P. Urban, Jr., K. M. Stoyanoff, J. Leung,
time, based on systematic review. Gastroenterology 142: 46–54, e42, quiz e30. and J. V. Weinstock. 2012. Heligmosomoides polygyrus bakeri induces tolerogenic
2. Ng, S. C., C. N. Bernstein, M. H. Vatn, P. L. Lakatos, E. V. Loftus, Jr., C. Tysk, dendritic cells that block colitis and prevent antigen-specific gut T cell responses. J.
C. O’Morain, B. Moum, and J. F. Colombel, Epidemiology and Natural History Immunol. 189: 2512–2520.
Task Force of the International Organization of Inflammatory Bowel Disease 28. Idoyaga, J., C. Fiorese, L. Zbytnuik, A. Lubkin, J. Miller, B. Malissen, D. Mucida,
(IOIBD). 2013. Geographical variability and environmental risk factors in in- M. Merad, and R. M. Steinman. 2013. Specialized role of migratory dendritic cells
flammatory bowel disease. Gut 62: 630–649. in peripheral tolerance induction. J. Clin. Invest. 123: 844–854.
3246 BRIEF REVIEWS: HELMINTHS REGULATE HOST IMMUNITY

29. Zhao, J., J. Wang, H. Wang, and M. Lai. 2013. Reg proteins and their roles in 2012. Therapeutic helminth infection of macaques with idiopathic chronic diar-
inflammation and cancer of the human digestive system. Adv. Clin. Chem. 61: rhea alters the inflammatory signature and mucosal microbiota of the colon. PLoS
153–173. Pathog. 8: e1003000.
30. Vaishnava, S., M. Yamamoto, K. M. Severson, K. A. Ruhn, X. Yu, O. Koren, 55. Leung, J. M., and P. Loke. 2013. A role for IL-22 in the relationship between
R. Ley, E. K. Wakeland, and L. V. Hooper. 2011. The antibacterial lectin intestinal helminths, gut microbiota and mucosal immunity. Int. J. Parasitol. 43:
RegIIIgamma promotes the spatial segregation of microbiota and host in the in- 253–257.
testine. Science 334: 255–258. 56. Broadhurst, M. J., J. M. Leung, V. Kashyap, J. M. McCune, U. Mahadevan,
31. van Ampting, M. T., L. M. Loonen, A. J. Schonewille, I. Konings, C. Vink, J. H. McKerrow, and P. Loke. 2010. IL-22+ CD4+ T cells are associated with
J. Iovanna, M. Chamaillard, J. Dekker, R. van der Meer, J. M. Wells, and therapeutic trichuris trichiura infection in an ulcerative colitis patient. Sci. Transl.
I. M. Bovee-Oudenhoven. 2012. Intestinally secreted C-type lectin Reg3b Med. 2: 60ra88.
attenuates salmonellosis but not listeriosis in mice. Infect. Immun. 80: 1115–1120. 57. Kron, M. A., A. Metwali, S. Vodanovic-Jankovic, and D. Elliott. 2013. Nematode
32. Bishnupuri, K. S., Q. Luo, S. K. Sainathan, K. Kikuchi, S. M. Sureban, asparaginyl-tRNA synthetase resolves intestinal inflammation in mice with T-cell
M. Sabarinathan, J. H. Gross, K. Aden, R. May, C. W. Houchen, S. Anant, and transfer colitis. Clin. Vaccine Immunol. 20: 276–281.
B. K. Dieckgraefe. 2010. Reg IV regulates normal intestinal and colorectal cancer 58. Knight, J. C. 2013. Genomic modulators of the immune response. Trends Genet.
cell susceptibility to radiation-induced apoptosis. Gastroenterology 138: 616–626, 29: 74–83.
626.e1–2. 59. Kurschus, F. C., S. Wörtge, and A. Waisman. 2011. Modeling a complex disease:
33. Sancho, D., and C. Reis e Sousa. 2013. Sensing of cell death by myeloid C-type multiple sclerosis. Adv. Immunol. 110: 111–137.
lectin receptors. Curr. Opin. Immunol. 25: 46–52. 60. Sewell, D., Z. Qing, E. Reinke, D. Elliot, J. Weinstock, M. Sandor, and Z. Fabry.
34. Dragicevic, A., T. Dzopalic, S. Vasilijic, D. Vucevic, S. Tomic, B. Bozic, and 2003. Immunomodulation of experimental autoimmune encephalomyelitis by
M. Colic. 2012. Signaling through Toll-like receptor 3 and Dectin-1 potentiates helminth ova immunization. Int. Immunol. 15: 59–69.
the capability of human monocyte-derived dendritic cells to promote T-helper 1 61. La Flamme, A. C., K. Ruddenklau, and B. T. Bäckström. 2003. Schistosomiasis
and T-helper 17 immune responses. Cytotherapy 14: 598–607. decreases central nervous system inflammation and alters the progression of ex-
35. Plato, A., J. A. Willment, and G. D. Brown. 2013. C-type lectin-like receptors of perimental autoimmune encephalomyelitis. Infect. Immun. 71: 4996–5004.
the dectin-1 cluster: ligands and signaling pathways. Int. Rev. Immunol. 32: 134– 62. Wilson, M. S., M. D. Taylor, M. T. O’Gorman, A. Balic, T. A. Barr, K. Filbey,
156. S. M. Anderton, and R. M. Maizels. 2010. Helminth-induced CD19+CD23hi
36. Kreider, T., R. M. Anthony, J. F. Urban, Jr., and W. C. Gause. 2007. Alternatively B cells modulate experimental allergic and autoimmune inflammation. Eur. J.
activated macrophages in helminth infections. Curr. Opin. Immunol. 19: 448–453. Immunol. 40: 1682–1696.

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


37. Reyes, J. L., and L. I. Terrazas. 2007. The divergent roles of alternatively activated 63. Walsh, K. P., M. T. Brady, C. M. Finlay, L. Boon, and K. H. Mills. 2009. In-
macrophages in helminthic infections. Parasite Immunol. 29: 609–619. fection with a helminth parasite attenuates autoimmunity through TGF-beta-
38. Smith, P., N. E. Mangan, C. M. Walsh, R. E. Fallon, A. N. McKenzie, N. van mediated suppression of Th17 and Th1 responses. J. Immunol. 183: 1577–1586.
Rooijen, and P. G. Fallon. 2007. Infection with a helminth parasite prevents ex- 64. Kuijk, L. M., E. J. Klaver, G. Kooij, S. M. van der Pol, P. Heijnen, S. C. Bruijns,
perimental colitis via a macrophage-mediated mechanism. J. Immunol. 178: 4557– H. Kringel, E. Pinelli, G. Kraal, H. E. de Vries, et al. 2012. Soluble helminth
4566. products suppress clinical signs in murine experimental autoimmune encephalo-
39. Schnoeller, C., S. Rausch, S. Pillai, A. Avagyan, B. M. Wittig, C. Loddenkemper, myelitis and differentially modulate human dendritic cell activation. Mol. Immu-
A. Hamann, E. Hamelmann, R. Lucius, and S. Hartmann. 2008. A helminth nol. 51: 210–218.
immunomodulator reduces allergic and inflammatory responses by induction of 65. Gruden-Movsesijan, A., N. Ilic, M. Mostarica-Stojkovic, S. Stosic-Grujicic,
IL-10-producing macrophages. J. Immunol. 180: 4265–4272. M. Milic, and L. Sofronic-Milosavljevic. 2010. Mechanisms of modulation of
40. Klotz, C., T. Ziegler, A. S. Figueiredo, S. Rausch, M. R. Hepworth, N. Obsivac, experimental autoimmune encephalomyelitis by chronic Trichinella spiralis infec-
C. Sers, R. Lang, P. Hammerstein, R. Lucius, and S. Hartmann. 2011. A helminth tion in Dark Agouti rats. Parasite Immunol. 32: 450–459.
immunomodulator exploits host signaling events to regulate cytokine production 66. Sofronic-Milosavljevic, L. J., I. Radovic, N. Ilic, I. Majstorovic, J. Cvetkovic, and
in macrophages. PLoS Pathog. 7: e1001248. A. Gruden-Movsesijan. 2013. Application of dendritic cells stimulated with
41. Johnston, M. J., A. Wang, M. E. Catarino, L. Ball, V. C. Phan, J. A. MacDonald, Trichinella spiralis excretory-secretory antigens alleviates experimental autoimmune
and D. M. McKay. 2010. Extracts of the rat tapeworm, Hymenolepis diminuta, encephalomyelitis. Med. Microbiol. Immunol. (Berl.) 202: 239–249.
suppress macrophage activation in vitro and alleviate chemically induced colitis in 67. Reyes, J. L., A. F. Espinoza-Jiménez, M. I. González, L. Verdin, and L. I. Terrazas.
mice. Infect. Immun. 78: 1364–1375. 2011. Taenia crassiceps infection abrogates experimental autoimmune encephalo-
42. Hunter, M. M., A. Wang, K. S. Parhar, M. J. Johnston, N. Van Rooijen, myelitis. Cell. Immunol. 267: 77–87.
P. L. Beck, and D. M. McKay. 2010. In vitro-derived alternatively activated 68. Terrazas, C. A., M. Alcántara-Hernández, L. Bonifaz, L. I. Terrazas, and
macrophages reduce colonic inflammation in mice. Gastroenterology 138: 1395– A. R. Satoskar. 2013. Helminth-excreted/secreted products are recognized by
1405. multiple receptors on DCs to block the TLR response and bias Th2 polarization in
43. Leung, G., A. Wang, M. Fernando, V. C. Phan, and D. M. McKay. 2013. Bone a cRAF dependent pathway. FASEB J. 27: 4547–4560.
marrow-derived alternatively activated macrophages reduce colitis without pro- 69. Correale, J., and M. F. Farez. 2011. The impact of parasite infections on the course
moting fibrosis: participation of IL-10. Am. J. Physiol. Gastrointest. Liver Physiol. of multiple sclerosis. J. Neuroimmunol. 233: 6–11.
304: G781–G792. 70. Correale, J., and M. F. Farez. 2013. Parasite infections in multiple sclerosis
44. Klaver, E. J., L. M. Kuijk, L. C. Laan, H. Kringel, S. J. van Vliet, G. Bouma, modulate immune responses through a retinoic acid-dependent pathway. J.
R. D. Cummings, G. Kraal, and I. van Die. 2013. Trichuris suis-induced modu- Immunol. 191: 3827–3837.
lation of human dendritic cell function is glycan-mediated. Int. J. Parasitol. 43: 71. Leiter, E. H., and A. Schile. 2013. Genetic and Pharmacologic Models for Type 1
191–200. Diabetes. Curr. Protoc. Mouse Biol. 3: 9–19.
45. Daniłowicz-Luebert, E., S. Steinfelder, A. A. K€uhl, G. Drozdenko, R. Lucius, 72. Cooke, A., P. Tonks, F. M. Jones, H. O’Shea, P. Hutchings, A. J. Fulford, and
M. Worm, E. Hamelmann, and S. Hartmann. 2013. A nematode immunomod- D. W. Dunne. 1999. Infection with Schistosoma mansoni prevents insulin de-
ulator suppresses grass pollen-specific allergic responses by controlling excessive pendent diabetes mellitus in non-obese diabetic mice. Parasite Immunol. 21: 169–
Th2 inflammation. Int. J. Parasitol. 43: 201–210. 176.
46. Rzepecka, J., I. Siebeke, J. C. Coltherd, D. E. Kean, C. N. Steiger, L. Al-Riyami, 73. Liu, Q., K. Sundar, P. K. Mishra, G. Mousavi, Z. Liu, A. Gaydo, F. Alem,
C. McSharry, M. M. Harnett, and W. Harnett. 2013. The helminth product, ES- D. Lagunoff, D. Bleich, and W. C. Gause. 2009. Helminth infection can reduce
62, protects against airway inflammation by resetting the Th cell phenotype. Int. J. insulitis and type 1 diabetes through CD25- and IL-10-independent mechanisms.
Parasitol. 43: 211–223. Infect. Immun. 77: 5347–5358.
47. Hartmann, W., Y. Brenz, M. T. Kingsley, I. Ajonina-Ekoti, N. W. Brattig, 74. Saunders, K. A., T. Raine, A. Cooke, and C. E. Lawrence. 2007. Inhibition of
E. Liebau, and M. Breloer. 2013. Nematode-derived proteins suppress prolifera- autoimmune type 1 diabetes by gastrointestinal helminth infection. Infect. Immun.
tion and cytokine production of antigen-specific T cells via induction of cell death. 75: 397–407.
PLoS ONE 8: e68380. 75. H€ ubner, M. P., Y. Shi, M. N. Torrero, E. Mueller, D. Larson, K. Soloviova,
48. Adisakwattana, P., S. P. Saunders, H. J. Nel, and P. G. Fallon. 2009. Helminth- F. Gondorf, A. Hoerauf, K. E. Killoran, J. T. Stocker, et al. 2012. Helminth
derived immunomodulatory molecules. Adv. Exp. Med. Biol. 666: 95–107. protection against autoimmune diabetes in nonobese diabetic mice is independent
49. Hewitson, J. P., J. R. Grainger, and R. M. Maizels. 2009. Helminth immuno- of a type 2 immune shift and requires TGF-b. J. Immunol. 188: 559–568.
regulation: the role of parasite secreted proteins in modulating host immunity. 76. Zaccone, P., O. Burton, N. Miller, F. M. Jones, D. W. Dunne, and A. Cooke.
Mol. Biochem. Parasitol. 167: 1–11. 2009. Schistosoma mansoni egg antigens induce Treg that participate in diabetes
50. Segura, M., Z. Su, C. Piccirillo, and M. M. Stevenson. 2007. Impairment of prevention in NOD mice. Eur. J. Immunol. 39: 1098–1107.
dendritic cell function by excretory-secretory products: a potential mechanism for 77. Lund, M. E., B. A. O’Brien, A. T. Hutchinson, M. W. Robinson, A. M. Simpson,
nematode-induced immunosuppression. Eur. J. Immunol. 37: 1887–1904. J. P. Dalton, and S. Donnelly. 2014. Secreted proteins from the helminth Fasciola
51. Taylor, B. C., C. Zaph, A. E. Troy, Y. Du, K. J. Guild, M. R. Comeau, and hepatica inhibit the initiation of autoreactive T cell responses and prevent diabetes
D. Artis. 2009. TSLP regulates intestinal immunity and inflammation in mouse in the NOD mouse. PLoS ONE 9: e86289.
models of helminth infection and colitis. J. Exp. Med. 206: 655–667. 78. Mishra, P. K., N. Patel, W. Wu, D. Bleich, and W. C. Gause. 2013. Prevention of
52. Strober, W. 2009. The multifaceted influence of the mucosal microflora on mu- type 1 diabetes through infection with an intestinal nematode parasite requires IL-
cosal dendritic cell responses. Immunity 31: 377–388. 10 in the absence of a Th2-type response. Mucosal Immunol. 6: 297–308.
53. Walk, S. T., A. M. Blum, S. A. Ewing, J. V. Weinstock, and V. B. Young. 2010. 79. Espinoza-Jiménez, A., I. Rivera-Montoya, R. Cárdenas-Arreola, L. Morán, and
Alteration of the murine gut microbiota during infection with the parasitic hel- L. I. Terrazas. 2010. Taenia crassiceps infection attenuates multiple low-dose
minth Heligmosomoides polygyrus. Inflamm. Bowel Dis. 16: 1841–1849. streptozotocin-induced diabetes. J. Biomed. Biotechnol. 2010: 850541.
54. Broadhurst, M. J., A. Ardeshir, B. Kanwar, J. Mirpuri, U. M. Gundra, 80. Osada, Y., S. Yamada, A. Nabeshima, Y. Yamagishi, K. Ishiwata, S. Nakae,
J. M. Leung, K. E. Wiens, I. Vujkovic-Cvijin, C. C. Kim, F. Yarovinsky, et al. K. Sudo, and T. Kanazawa. 2013. Heligmosomoides polygyrus infection reduces
The Journal of Immunology 3247

severity of type 1 diabetes induced by multiple low-dose streptozotocin in mice via 94. Kitagaki, K., T. R. Businga, D. Racila, D. E. Elliott, J. V. Weinstock, and
STAT6- and IL-10-independent mechanisms. Exp. Parasitol. 135: 388–396. J. N. Kline. 2006. Intestinal helminths protect in a murine model of asthma. J.
81. Brand, D. D., A. H. Kang, and E. F. Rosloniec. 2003. Immunopathogenesis of Immunol. 177: 1628–1635.
collagen arthritis. Springer Semin. Immunopathol. 25: 3–18. 95. Wilson, M. S., M. D. Taylor, A. Balic, C. A. Finney, J. R. Lamb, and
82. Osada, Y., S. Shimizu, T. Kumagai, S. Yamada, and T. Kanazawa. 2009. Schis- R. M. Maizels. 2005. Suppression of allergic airway inflammation by helminth-
tosoma mansoni infection reduces severity of collagen-induced arthritis via down- induced regulatory T cells. J. Exp. Med. 202: 1199–1212.
regulation of pro-inflammatory mediators. Int. J. Parasitol. 39: 457–464. 96. McSorley, H. J., M. T. O’Gorman, N. Blair, T. E. Sutherland, K. J. Filbey, and
83. He, Y., J. Li, W. Zhuang, L. Yin, C. Chen, J. Li, F. Chi, Y. Bai, and X. P. Chen. R. M. Maizels. 2012. Suppression of type 2 immunity and allergic airway in-
2010. The inhibitory effect against collagen-induced arthritis by Schistosoma flammation by secreted products of the helminth Heligmosomoides polygyrus. Eur. J.
japonicum infection is infection stage-dependent. BMC Immunol. 11: 28. Immunol. 42: 2667–2682.
84. Song, X., J. Shen, H. Wen, Z. Zhong, Q. Luo, D. Chu, Y. Qi, Y. Xu, and W. Wei. 97. Smits, H. H., H. Hammad, M. van Nimwegen, T. Soullie, M. A. Willart,
2011. Impact of Schistosoma japonicum infection on collagen-induced arthritis in E. Lievers, J. Kadouch, M. Kool, J. Kos-van Oosterhoud, A. M. Deelder, et al.
DBA/1 mice: a murine model of human rheumatoid arthritis. PLoS ONE 6: e23453. 2007. Protective effect of Schistosoma mansoni infection on allergic airway in-
85. Sun, X., Y. H. Liu, Z. Y. Lv, L. L. Yang, S. M. Hu, H. Q. Zheng, W. Hu, flammation depends on the intensity and chronicity of infection. J. Allergy Clin.
J. P. Cao, M. Q. Fung, and Z. D. Wu. 2010. rSj16, a recombinant protein of Immunol. 120: 932–940.
Schistosoma japonicum-derived molecule, reduces severity of the complete Freund’s 98. Pacı́fico, L. G., F. A. Marinho, C. T. Fonseca, M. M. Barsante, V. Pinho,
adjuvant-induced adjuvant arthritis in rats’ model. Parasite Immunol. 32: 739–748. P. A. Sales-Junior, L. S. Cardoso, M. I. Araújo, E. M. Carvalho, G. D. Cassali,
86. McInnes, I. B., B. P. Leung, M. Harnett, J. A. Gracie, F. Y. Liew, and W. Harnett. et al. 2009. Schistosoma mansoni antigens modulate experimental allergic asthma in
2003. A novel therapeutic approach targeting articular inflammation using the a murine model: a major role for CD4+ CD25+ Foxp3+ T cells independent of
filarial nematode-derived phosphorylcholine-containing glycoprotein ES-62. J. interleukin-10. Infect. Immun. 77: 98–107.
Immunol. 171: 2127–2133. 99. Layland, L. E., K. Straubinger, M. Ritter, E. Loffredo-Verde, H. Garn,
87. Pineda, M. A., M. A. McGrath, P. C. Smith, L. Al-Riyami, J. Rzepecka, T. Sparwasser, and C. Prazeres da Costa. 2013. Schistosoma mansoni-mediated
J. A. Gracie, W. Harnett, and M. M. Harnett. 2012. The parasitic helminth suppression of allergic airway inflammation requires patency and Foxp3+ Treg
product ES-62 suppresses pathogenesis in collagen-induced arthritis by targeting cells. PLoS Negl. Trop. Dis. 7: e2379.
the interleukin-17-producing cellular network at multiple sites. Arthritis Rheum. 100. Liu, P., J. Li, X. Yang, Y. Shen, Y. Zhu, S. Wang, Z. Wu, X. Liu, G. An, W. Ji,
64: 3168–3178. et al. 2010. Helminth infection inhibits airway allergic reaction and dendritic cells
88. Al-Riyami, L., M. A. Pineda, J. Rzepecka, J. K. Huggan, A. I. Khalaf, are involved in the modulation process. Parasite Immunol. 32: 57–66.

Downloaded from http://www.jimmunol.org/ by guest on April 4, 2020


C. J. Suckling, F. J. Scott, D. T. Rodgers, M. M. Harnett, and W. Harnett. 2013. 101. Park, H. K., M. K. Cho, S. H. Choi, Y. S. Kim, and H. S. Yu. 2011. Trichinella
Designing anti-inflammatory drugs from parasitic worms: a synthetic small mol- spiralis: infection reduces airway allergic inflammation in mice. Exp. Parasitol. 127:
ecule analogue of the Acanthocheilonema viteae product ES-62 prevents develop- 539–544.
ment of collagen-induced arthritis. J. Med. Chem. 56: 9982–10002. 102. Aranzamendi, C., A. de Bruin, R. Kuiper, C. J. Boog, W. van Eden, V. Rutten,
89. Carranza, F., C. R. Falcón, N. Nuñez, C. Knubel, S. G. Correa, I. Bianco, and E. Pinelli. 2013. Protection against allergic airway inflammation during the
M. Maccioni, R. Fretes, M. F. Triquell, C. C. Motrán, and L. Cervi. 2012. chronic and acute phases of Trichinella spiralis infection. Clin. Exp. Allergy 43:
Helminth antigens enable CpG-activated dendritic cells to inhibit the symptoms of 103–115.
collagen-induced arthritis through Foxp3+ regulatory T cells. PLoS ONE 7: 103. Amu, S., S. P. Saunders, M. Kronenberg, N. E. Mangan, A. Atzberger, and
e40356. P. G. Fallon. 2010. Regulatory B cells prevent and reverse allergic airway in-
90. Shi, M., A. Wang, D. Prescott, C. C. Waterhouse, S. Zhang, J. J. McDougall, flammation via FoxP3-positive T regulatory cells in a murine model. J. Allergy
K. A. Sharkey, and D. M. McKay. 2011. Infection with an intestinal helminth Clin. Immunol. 125: 1114–1124, e8.
parasite reduces Freund’s complete adjuvant-induced monoarthritis in mice. Ar- 104. Melendez, A. J., M. M. Harnett, P. N. Pushparaj, W. S. Wong, H. K. Tay,
thritis Rheum. 63: 434–444. C. P. McSharry, and W. Harnett. 2007. Inhibition of Fc epsilon RI-mediated mast
91. Graepel, R., G. Leung, A. Wang, M. Villemaire, F. R. Jirik, K. A. Sharkey, cell responses by ES-62, a product of parasitic filarial nematodes. Nat. Med. 13:
J. J. McDougall, and D. M. McKay. 2013. Murine autoimmune arthritis is ex- 1375–1381.
aggerated by infection with the rat tapeworm, Hymenolepis diminuta. Int. J. Par- 105. Fumagalli, M., U. Pozzoli, R. Cagliani, G. P. Comi, S. Riva, M. Clerici,
asitol. 43: 593–601. N. Bresolin, and M. Sironi. 2009. Parasites represent a major selective force for
92. Amoah, A. S., D. A. Boakye, R. van Ree, and M. Yazdanbakhsh. 2014. Parasitic interleukin genes and shape the genetic predisposition to autoimmune conditions.
worms and allergies in childhood: insights from population studies 2008-2013. J. Exp. Med. 206: 1395–1408.
Pediatr. Allergy Immunol. 25: 208–217. 106. Fumagalli, M., U. Pozzoli, R. Cagliani, G. P. Comi, N. Bresolin, M. Clerici, and
93. Zosky, G. R., and P. D. Sly. 2007. Animal models of asthma. Clin. Exp. Allergy 37: M. Sironi. 2010. The landscape of human genes involved in the immune response
973–988. to parasitic worms. BMC Evol. Biol. 10: 264.

You might also like