You are on page 1of 9

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/26827642

Regulation of pathogenesis and immunity in


helminth infections

Article in Journal of Experimental Medicine · September 2009


DOI: 10.1084/jem.20091903 · Source: PubMed

CITATIONS READS

141 95

5 authors, including:

Maria Yazdanbakhsh Thomas A Wynn


Leiden University Medical Centre National Institutes of Health
339 PUBLICATIONS 12,451 CITATIONS 331 PUBLICATIONS 30,093 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Regulatory B cells and their activity in patients with allergic asthma View project

All content following this page was uploaded by Thomas A Wynn on 24 July 2014.

The user has requested enhancement of the downloaded file.


Meeting Review

Regulation of pathogenesis and immunity


in helminth infections
Rick M. Maizels, Edward J. Pearce, David Artis, Maria Yazdanbakhsh, and Thomas A. Wynn

Helminths are multicellular eukaryotic parasites that infect over one quarter
of the world’s population. Through coevolution with the human immune

IMAGE COURTESY OF CONSTANCE FINNEY.


system, these organisms have learned to exploit immunoregulatory pathways,
resulting in asymptomatic tolerance of infections in many individuals. When
infections and the resulting immune responses become dysregulated, however,
acute and chronic pathologies often develop. A recent international meeting
focused on how these parasites modulate host immunity and how control of
parasitic and immunopathological disease might be achieved.
The Journal of Experimental Medicine

It is widely accepted that helminths How the human immune system


and their antigens inherently induce T fights off—or learns to live with—
helper (Th) 2 responses, and it is likely worm infections has been extensively The intestinal helminth H. polygyrus.
that Th2 immunity evolved in response studied, but many questions remain
to infections with these parasites. In unanswered. On the host side, the co-
February 2009, scientists from 32 coun- ordinate functions of multiple cell Another Th2-inducing cytokine,
tries assembled in Tahoe City, CA for types, including phagocytes, granulo- thymic stromal lymphopoietin (TSLP),
the Keystone symposium on Patho- cytes, B cells, Th2 cells, and regulatory may also be dispensable for generating
genesis and Immune Regulation in T (T reg) cells, dictate the outcome Th2 responses against certain helminth
Helminth Infections (February 1–5, of infection. infections. In noninfectious settings,
2009), organized by Rick Maizels, such as allergy, Th2 responses are
Maria Yazdanbakhsh, and Thomas Wynn, Getting things started: Th2 cells largely TSLP dependent. But in the ab-
to discuss how immune responses de- and beyond sence of a functional TSLP receptor,
velop in response to helminth infec- IL-4–producing Th2 cells are perhaps mice infected with Heligmosomoides
tions and how these infections are (or the most extensively studied cells in the polygyrus, N. brasiliensis, or Schistosoma
are not) controlled. The discussions context of worm infections, and gener- mansoni (Fig. 1). still generated strong
provided new insights into the cellular ating an effective Th2 response has Th2 responses (Nicola Harris, Lau­
players, cytokines, and effector mole- long been thought to require the early sanne, Switzerland; Massacand et al.,
cules that help initiate, and subse- production of interleukin (IL)-4. Con- 2009; Ramalingam et al., 2009). This
quently limit, immune responses during trary to this idea, however, Graham Le rule does not hold true for all worm
helminth infection. Gros (Wellington, New Zealand) infections, however, as TSLP is essen-
R.M.M. is at Centre for Immunity, Infection and showed that neither IL-4 nor signal tial for the development of Th2 re-
Evolution, and Institute of Immunology and Infection transducer and activator of transcrip- sponses against Trichuris muris (Taylor
Research, University of Edinburgh, Edinburgh, tion (STAT)-6, a key component of et al., 2009a). A key difference between
Scotland, UK.
the IL-4R signaling pathway, was es- these parasites appears to be the ability
E.J.P. is at Trudeau Institute, Saranac Lake, NY 12983. sential for the development of a Th2 of H. polygyrus and N. brasiliensis, but
response in vivo. Activation of an IL-4– not T. muris, to inhibit the production
D.A. is at Department of Pathobiology, School of
Veterinary Medicine, University of Pennsylvania,
linked GFP reporter construct was of inflammatory cytokines, such as IL-6,
Philadelphia, PA 19104. intact in STAT-6–deficient mice in- IL-12, and TNF, from dendritic cells
fected with Nippostrongylus brasiliensis (DCs) in response to Toll-like receptor
M.Y. is at Department of Parasitology, Leiden (King et al., 2008; van Panhuys et al., (TLR) ligation. T. muris may thus re-
University Medical Center, Leiden, Netherlands.
2008), suggesting that other factors, quire TSLP to help restrain IL-12 pro-
T.A.W. is at Immunopathogenesis Section, Laboratory whether host- or parasite-derived, are duction before a Th2 response can
of Parasitic Diseases, National Institute of Allergy and critical for Th2 polarization in vivo. develop, whereas other parasites inhibit
Infectious Diseases, National Institutes of Health,
Bethesda, MD 20892.
© 2009 Maizels et al.  This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the
CORRESPONDENCE first six months after the publication date (see http://www.jem.org/misc/terms.shtml). After six months it is available under a Creative Commons
R.M.M.: rick.maizels@ed.ac.uk License (Attribution–Noncommercial–Share Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/by-nc-sa/3.0/).

The Rockefeller University Press  $30.00


J. Exp. Med. Vol. 206 No. 10  2059-2066 2059
www.jem.org/cgi/doi/10.1084/jem.20091903
cytokine production directly, thereby about the precise definition of a Th2 are required to initiate the host re-
circumventing this pathway. cell. Locksley argued that conventional sponse. New data from Markus Mohrs
Where and how Th2 cells are gen- Th2 cells may function primarily within (Saranac Lake, NY) revealed that the
erated has been the subject of intense tissues to mediate classical allergic-like Th2-inducing potential of schistosome
scrutiny. Using IL-4-GFP (“4get”) re- inflammation, such as eosinophil re- eggs is due in part to the action of
porter mice to track cells that produce cruitment, whereas Tfh cells function omega-1, a glycoprotein with ribonu-
IL-4 in response to infection, multiple in the follicles to deliver help to B cells. clease activity that is released from eggs
groups have found that follicular T Pearce offered further perspective on under physiological conditions (Everts
helper (Tfh) cells are the predominant the Th2 population in Schistosoma in- et al., 2009). Purified omega-1 induced
IL-4–producing cells in responding fection by showing that Th2 cells lose strong Th2 responses in mice, even in
lymph nodes (Richard Locksley, San responsiveness as chronic disease devel- those lacking the IL-4 receptor. Omega-1
Francisco, CA and Edward Pearce, Sa- ops, a process mediated by increased also inhibited DC activation in re-
ranac Lake, NY; Reinhardt et al., 2009; expression of GRAIL, an E3 ubiquitin sponse to lipopolysaccharide, suggest-
Zaretsky et al., 2009). Similar findings ligase implicated in the development of ing that the egg protein acts at the initial
have also been reported by Markus T cell anergy (Taylor et al., 2009b). stages of response (Steinfelder et al.,
Mohrs (King and Mohrs, 2009). This These and other studies have taught 2009). Production of omega-1 likely
may not seem surprising, considering us a great deal about the host require- helps sustain infection, as the subse-
the well-established role of IL-4 in B ments for generating Th2 responses quent Th2 response stimulates the for-
cell activation and Ig class switching, against helminths. But far less is known mation of protective granulomas around
but it nevertheless raised questions about what components of the parasites the eggs. The hunt is now on to iden-
tify the cellular receptor for omega-1.
Like most helminth infections,
schistosomiasis elicits a predominant
CD4+ Th2 cell response, and mice de-
pend on this response to survive infec-
tion. In the absence of IL-4, mice
develop hepatotoxicity, endotoxemia,
and severe cachexia, which together
contribute to the death of the animal.
However, some strains of mice, such as
CBA, are more prone to developing
Th1 and Th17 responses, and thus de-
velop severe pathology in response to
S. mansoni infection (Miguel Stadecker
and Mara Shainheit, Boston, MA). DCs
from CBA mice produced more IL-
12p40 and IL-6 in response to live
schistosome eggs than did DCs from
C57BL/6 mice. CBA DCs also stimu-
lated stronger IL-17 production from
transgenic T cells specific for the egg
antigen Sm-p40. In vitro experiments
suggested that the increased Th17 re-
sponse requires both IL-1 and IL-23
(Rutitzky et al., 2008) and that the
Th17 response is responsible for the
severe egg-induced inflammatory re-
sponse seen in infected CBA mice
(Shainheit et al., 2008). Co-infection
with the intestinal nematode H. poly-
gyrus decreased IL-17 and interferon
Figure 1.  Helminth infections. A summary of the major helminth infections of humans and their
(IFN)- production in CBA mice, per-
mouse model counterparts. Estimates of human infection numbers are taken from Hotez et al. haps helping to explain the low inci-
(2008). Note that the schematic does not include highly prevalent helminth infections such as Asca- dence of autoimmune diseases in regions
ris lumbricoides (807 million estimated infections), other important human parasites, or the promi- where helminth infections are endemic
nent veterinary helminth organisms. (Bazzone et al., 2008).

2060 immunoregulatory helminths | Maizels et al.


Meeting Review

Keeping potentially dangerous Th1 hances parasite killing, illustrating the splenic disease (Tom Wynn, Bethesda,
responses at bay in T. muris infection opposing roles of these pathways in MD). The protective action of AAMs
depends on activation of the NF-B anti-helminth immunity. depended on expression of arginase-1
signaling pathway in intestinal epithelial T reg cells also suppress host im- (Arg-1), as mice whose macrophages
cells (IECs), according to David Artis mune responses in humans, presumably lack Arg-1 failed to suppress Th2 re-
(Philadelphia, PA). In mice with an allowing parasites to establish chronic sponses and egg-induced inflammation.
IEC-specific ablation of the classical infections (Maizels and Yazdanbakhsh, As a result, the mice developed fibrosis
NF-B pathway, DCs produced excess 2003). Indeed, schistosome-infected in- (Pesce et al., 2009a). The protective
inflammatory cytokines leading to the dividuals in Gabon had increased num- effect of AAMs is likely to involve
development of a nonprotective Th1 bers of circulating T reg cells compared the arginase-dependent depletion of
response (Zaph et al., 2007). Normally, with their uninfected neighbors (Maria L-arginine, which is required to main-
the DC-triggered induction of a Th2 Yazdanbakhsh, Leiden, Netherlands tain T cell proliferation and Th2
response depends on the NF-B– and Ayola Akim Adegnika, Lambaréné, responses (Pesce et al., 2009a).
dependent production of TSLP and Gabon). And helminth-induced T reg Consistently, macrophages from mice
IL-25 from IECs (Zaph et al., 2008; cells may have more potent suppressive lacking the cationic amino acid trans-
Taylor et al., 2009a). activity than those from healthy indi- porter 2 (Cat2), which regulates L-argi-
viduals. T reg cells from geohelminth- nine transport into cells, also had
Applying the brakes: T reg cells infected individuals in Indonesia, for increased Arg1 activity and reduced
Generating a Th2 response to parasite example, were more effective at sup- Th2 activity. Surprisingly, however,
infections is essential, but controlling pressing proliferation and IFN- pro- Cat2-deficient mice developed severe
that response is equally imperative. duction by effector T cells in response pathology despite their reduced Th2
Some of this control is provided by to malaria antigens and BCG than responses, perhaps owing to enhanced
T reg cells, which were shown to dampen T reg cells from healthy individuals Arg1 activity in fibroblasts (Thompson
Th2 responses during H. polygyrus in- (Yazdanbakhsh and Taniawati Supali, et al., 2008).
fection (Rick Maizels, Edinburgh, Jakarta, Indonesia). Tom Nutman Another product of AAMs, resistin-
Scotland, UK; Wilson et al., 2005). In (Bethesda, MD) also reported dimin- like molecule- (RELM/Fizz1/Retnla),
this model, the parasite itself drives the ished production of cytokines in re- helps to control inflammation in re-
differentiation of T reg cells from naive sponse to malaria antigens in peripheral sponse to S. mansoni. RELM can
CD4+ T cells by secreting a product blood cells from helminth/malaria also be produced by other cell types, as
that binds to host TGF- receptors. coinfected individuals. This inhibition macrophages, eosinophils, and epithe-
Inhibiting this interaction in vivo was driven primarily by the suppressive lial cells produced the molecule after
decreased the intensity of infection, cytokine IL-10 (Metenou et al., 2009) exposure to S. mansoni eggs in the lung
presumably as a result of more effective (Fig. 2; Meera Nair, Philadelphia, PA).
Th2 responses, illustrating the evolu- Maintaining balance: alternatively Mice lacking RELM developed se-
tionary advantage of this pathway for activated macrophages vere egg-induced inflammation in the
the parasite. Robust Th2 responses are required to lung and liver that was associated with
Induction of T reg cells is also clear helminth infections, but Th2 re- an enhanced Th2 response and could
needed for Litomosoides sigmodontis to sponses that overshoot can be danger- be reversed by treating the mice with
establish long-lasting infections. When ous. Although Th1 and Th17 responses recombinant RELM (Nair et al.,
T reg cells were depleted in susceptible develop in response to acute S. mansoni 2009; Pesce et al., 2009b).
mouse strains, infection was cleared infections in most mouse strains, they The dominant cellular sources of
(Nienke van der Werf and Matthew quickly wane and are replaced by Th2 RELM may depend on the stimulus
Taylor, Edinburgh, Scotland, UK; responses. Although this transition is and route of exposure. For example, af-
Taylor et al., 2009). In chronic infection, initially beneficial to the host, persistent ter S. mansoni infection, or during pri-
subsequent reactivation of hyporespon- Th2 responses can contribute to liver mary pulmonary granuloma formation,
sive Th2 cells, which express high levels pathology and hepatosplenic disease RELM was predominantly produced
of GITR, CTLA-4, and PD-1, required (Wynn, 2007). by eosinophils rather than macrophages
CTLA-4 blockade or co-stimulation of Prior studies had suggested that (Tom Wynn, Bethesda, MD; Pesce et al.,
the cells through GITR. Indeed, GITR alternatively activated macrophages 2009b). The role of AAM-derived
ligation was essential for the develop- (AAMs), which are triggered by Th2 RELM should thus be clarified
ment of Th2 responses during this in- responses, promote pathology during through studies with cell-specific dele-
fection. Recent data indicates that chronic infection (Wynn, 2008). But tions of Relm. A related molecule,
GITR also promotes the generation of more recent data show that AAMs ac- RELM, was shown to be required for
Th2 responses during the initial im- tually help to dampen immune re- expulsion of N. brasiliensis and H. poly-
mune-priming stage of infection and sponses in schistosomiasis and to inhibit gyrus, which both reside in the intestinal
that blocking the PD-1 pathway en- the development of severe hepato- lumen, but was not required for expulsion

JEM VOL. 206, September 28, 2009 2061


of T. spiralis, which lives within epithe- parasite killing. Given that helminths which MHC class II expression was re-
lial cells (De’Broski Herbert, Cincin- cause considerable damage while mi- stricted to DCs. These mice could not
nati, OH). RELM appears to inhibit grating through tissues, particularly the generate protective Th2 responses to
the ability of worms to feed on host tis- gut and lung, Allen and colleagues pro- T. muris, indicating the need for at least
sues resulting in reduced ATP content posed a model in which the immune one other antigen-presenting cell type.
and worm fecundity. system repairs helminth-induced dam- Using IL-4-GFP (“4get”) reporter
AAMs may also have a hand in the age more rapidly after secondary worm mice, they spotted IL-4–producing,
direct killing of parasites, as clodronate exposure in an IL-4/IL-13–dependent MHC class II+ basophils in the draining
liposome-mediated depletion of AAMs manner, accelerating the return of lymph nodes. When these cells were
dramatically inhibited killing of Brugia homeostasis to critical tissues (Loke depleted, Th2 responses were impaired.
malayi in mice (Judith Allen, Edinburgh, et al., 2007; Mylonas et al., 2009). Basophil responses were boosted by the
Scotland, UK). However, parasite kill- Whether helminth-elicited AAMs cytokine TSLP, which was produced
ing was also impaired in mice receiving play a role in the immune-regulation primarily by intestinal epithelial cells,
PBS-loaded liposomes or latex beads, in models of chronic inflammation and both cell types were required for an
which compromise macrophage func- awaits further study. optimal Th2 response (Perrigoue et al.,
tion without reducing cell numbers or 2009). The effects of basophils on Th2
the expression of AAM-associated genes New cells on the scene: basophils induction appear to be age-dependent,
(Arg1, RELM, and Ym1). This sug- Basophils are newly recognized players according to Padraic Fallon (Dublin,
gests that macrophages are critical for in the immune response to helminth Ireland), who showed that impaired ba-
parasite killing, but that alternative acti- infections. These cells, along with eo- sophil function in juvenile mice (<21 d
vation may not be required. Macro- sinophils, are known to produce IL-4 of age) resulted in insufficient IL-4 pro-
phage activation by Th2 cytokines during allergic responses and certain duction, poor Th2 generation, and an
might instead function primarily to reg- helminth infections (Fig. 3), but inability to expel N. brasiliensis.
ulate immune responses and to facilitate whether these cells were essential for
tissue repair rather than to promote clearing helminths was not clear. The B cells, antibodies, and symbionts
critical importance of these cells was B cells and antibodies are essential in
demonstrated in part by Locksley, who combatting some, but not all, helminth
showed that allergic responses and infections. Bill Gause (Newark, NJ)
worm expulsion (N. brasiliensis) did not demonstrated that both macrophages
rely on IL-4 production by T cells, as and B cells were required to prevent
these functions were intact in mice H. polygyrus from invading gut tissues
whose T cells lacked CD4+ IL-4 and within the first several days of a mem-
IL-13. Parasite expulsion was also intact ory response. But B cells were not re-
in mice lacking mast cells (KitW-sh) or quired for effective expulsion of N.
eosinophils (Gata-1 mutant). But baso- brasiliensis, suggesting that the impor-
phil depletion (using anti-MAR-1 anti- tance of B cells depends on the infect-
bodies) hindered worm clearance. The ing parasite.
ability of basophils to produce IL-4 is High levels of parasite-specific
likely to be critical during both primary IgE correlate with resistance to dis-
and secondary responses. Indeed, al- ease induced by S. mansoni infection
though schistosome-infected mice lack- (Butterworth et al., 1992), although
ing IL-4 and IL-13 succumbed to what triggers IgE production and how
infection, provision of IL-4 by non- these antibodies protect the host remain
T cells was sufficient to rescue them. And elusive. The protective role of IgE was
depleting basophils from immunodefi- studied in ongoing field studies, which
cient mice that had been injected with showed that repeated treatment with
IL-4/13-deficient lymphocytes and then the drug praziquantel over 3–6 yr ac-
infected with N. brasiliensis attenuated eo- celerated the development of resistance
Figure 2.  Expression of RELM proteins sinophil recruitment and worm expulsion to reinfection. Resistance correlated
after exposure to helminth parasites. RELM (David Voehringer, Munich, Germany; with increased levels of antigen-specific
(green) is expressed by mannose receptor–
Ohnmacht and Voehringer, 2009). IgE and increased expression of CD23,
positive cells (red) in the lungs of C57BL/6 mice
challenged i.v. with S. mansoni eggs (A), and
Basophils were also found in the the low-affinity IgE receptor, on B cells
RELM is expressed by goblet cells of the gas- draining lymph nodes of mice infected (Dan Colley, Athens, GA; Mwinzi
trointestinal tract in C57BL/6 mice infected orally with T. muris. The Artis group set out et al., 2009).
with T. muris eggs (B). Image courtesy of Meera to ask which antigen-presenting cells Some helminths, such as the filarial
Nair and David Artis, University of Pennsylvania. were critical in this model using mice in parasites, are infected with symbiotic

2062 immunoregulatory helminths | Maizels et al.


Meeting Review

Wolbachia bacteria, which has led to infections may contribute to a decreased pression of FoxP3 in naive NOD T
therapeutic use of doxycycline antibiot- incidence of type I diabetes. Illustrating cells (Zaccone et al., 2009), suggesting
ics to eliminate both endosymbionts this, Marc Hubner and Edward Mitre that inducible T reg cells may help pre-
and the worms they depend on. Achim (Bethesda, MD) reported that infection vent disease in this system.
Hoerauf (Bonn, Germany) showed that with L. sigmodontis or injection of Allergic responses are also muted by
TLR-dependent neutrophilic responses L. sigmodontis antigens prevented diabe- helminth infections (Maizels, 2005).
in filarial infections are stimulated by tes in NOD mice (Hübner et al., 2009). Exposure to the intestinal worm
Wolbachia rather than the worm itself. Protection was associated with increased N. brasiliensis or worm extracts decreased
Growth of tissue-dwelling filarial para- Th2 responses and T reg cell numbers, allergen-induced airway inflammation
sites also appears to be enhanced by an- rather than decreased pathogenic Th17 in mice sensitized to ovalbumin (OVA),
giogenic factors such as CCL17 (TARC) responses. Somewhat surprisingly, pro- including decreased eosinophilia, air-
and VEGF-A, and the latter was pro- tection did not require the signature way hyperreactivity, and OVA-specific
moted by the symbiotic bacteria. Th2 cytokine IL-4. Infection with IgG1 and IgE production (Klaus Erb,
S. mansoni also protected against diabe- Germany). The reduction in airway in-
Worm influences on allergy, tes in NOD mice, as discussed by Anne flammation was attributed to the induc-
autoimmunity, and vaccines Cooke (Cambridge, England, UK). tion of T reg cells and depended on
Infection with helminths can be benefi- CD25+ T reg cells played a crucial role IL-10 production. Although somewhat
cial to the host, as the Th2 and T reg in this model, as transfer of CD25+ cells counterintuitive, the protection against
cell responses that develop in response from infected mice protected against Th2-type allergic responses went hand-
to helminths can suppress allergic and disease. And in vitro studies demon- in-hand with increased worm-specific
autoimmune responses (Wilson and strated that schistosome egg antigens Th2 responses, which might have been
Maizels, 2004). For example, helminth triggered the TGF-–dependent ex- predicted to worsen the allergic

Figure 3.  Cellular interactions in the immune response to helminths. Helminths are complex eukaryotic pathogens with multiple life stages that
can affect different tissues within the parasitized host. Helminth-derived antigens can drain directly to secondary lymphoid organs or interact with cells
at the site of infection, including DCs (1) or epithelial cells (2). Many helminth antigens inhibit the production of proinflammatory cytokines by DCs (3),
and trigger the production of cytokines such as TSLP from epithelial cells, which inhibits IL-12 production by DCs. Antigen-carrying DCs activate naive
Th cells (4), which proliferate and differentiate into precursor Th2 cells (5). pTh2 cells can then become Th2 cells (6) or Tfh cells (7). Th2 cells release various
cytokines, including IL-5, which drives eosinophilia (8), and IL-4/-13, which stimulate AAMs (9). AAMs in turn produce molecules such as arginase-1 and
YM-1, which dampen Th2 responses. Tfh produce IL-4 and provide help to B cells for IgG1 and IgE production (10). During the early response to helminth
infections, MHC class II–expressing basophils (11) enter the reactive secondary lymphoid organs and may help to polarize the Th2 response. IgE can then
activate basophils (12), which in turn produce cytokines that activate alternative macrophages. Helminth infections can also promote the development of
T reg cell responses (13).

JEM VOL. 206, September 28, 2009 2063


response. How N. brasiliensis infection with Crohn’s disease and multiple scle- testinal helminth infections are endemic,
inhibits allergic responses without com- rosis. In an independent study, infec- efforts to eliminate or broadly control
promising helminth-specific Th2 immu­ tion with the hookworm Necator helminth infections may be required to
nity is not yet known. americanus was shown to reduce the maximize vaccine efficacy. And large-
Echoing the critical role of IL-10 in symptoms of Crohn’s disease (Danielle scale studies are needed to assess the full
suppressing inflammation, H. polygyrus Smyth, Brisbane, Australia; Croese et al., impact of helminth infections on the
infection of IL-10–deficient mice re- 2006). This group also found that secre- clinical course of coendemic infections,
lieved the spontaneous colonic inflam- tory antigens from the dog hookworm as well as inflammatory conditions such
mation that normally develops in these Ancylostoma caninum alleviated dextran as allergy and autoimmunity.
mice, according to Joel Weinstock sulfate sodium–induced colitis in mice Finally, there is an urgent agenda to
(Boston, MA). Turning to a mouse with reduced production of Th1 and develop protective anti-helminth vac-
model of OVA-induced colitis, Wein- Th17 cytokines, suggesting that a bene- cines, which are conspicuously absent
stock showed that H. polygyrus infection ficial effect might be achieved without from the world’s public health arma-
reduced colitis with fewer OVA-spe- active infection. mentarium. Peter Hotez (Washington,
cific Th1 and Th17 cells accumulating An excellent example of a helminth- DC) outlined progress made using the
in the lamina propria (Elliott et al., derived therapeutic was presented by most advanced vaccines for human
2008). In this model, TGF-–produc- Billy Harnett (Glasgow, Scotland, UK) hookworm, including Na-ASP-2,
ing cells expressing TLR4 also sup- on the filarial nematode protein ES-62. Na-GST-1, and Na-APR-1 (Loukas
pressed inflammation in response to This secreted protease bears crucial et al., 2006), with the latest updates on
lipopolysaccharide. phosphorylcholine sidechains that in- trials in endemic areas. The immuno­
Whether the protective effect of teract with B cells, macrophages, mast logical responses in vaccinated sub-
helminth infections in mice can be ex- cells, and DCs, and modulate TLR- jects will also be invaluable in
tended to humans remains to be dem- linked signaling pathways (Harnett and identifying beneficial and protective
onstrated on a broad scale. However, an Harnett, 2008). ES-62 can suppress im- responses and distinguishing them
impressive example of the power of mune responsiveness in vivo, in both from the regulatory responses induced
these infections to suppress autoimmu- collagen-induced arthritis and OVA- by helminth parasites.
nity was provided by P’ng Loke (San specific allergic airway inflammation.
Francisco, CA), who described a patient These studies highlight the poten- Concluding remarks
suffering from ulcerative colitis who re- tially beneficial side effects of helminth These studies have revealed ground-
lieved his intestinal inflammation by in- infection, but there is also a downside. breaking new findings about the biology
fecting himself with the intestinal worm By the same means that helminth in- of helminth infections and how mam-
Trichuris trichiura. The cytokine IL-22, fections benefit the host—i.e., by malian hosts respond to them, providing
which was expressed in a distinct popu- dampening potentially damaging in- a much clearer conceptual grasp of the
lation of CD4+ T cells in the lamina flammation—they also blunt responses balance between the immune reactivity
propria of this patient, may have helped to vaccines. Children infected with hel- and regulation that controls the outcome
control inflammation. Similarly, CD4+ minths, for example, generated rela- of helminth infections. Throughout the
T cell–derived IL-22 protects against tively poor multifunctional T cell meeting, it was clear that laboratory
inflammatory bowel disease in mice responses (coproduction of IL-2, TNF, models and field studies shared common
(Zenewicz et al., 2008). and IFN-) when vaccinated against in- themes, and it was gratifying to see labo-
A study of hookworm-infected in- fluenza (Yazdanbakhsh, Leiden, Neth- ratory findings being translated into
dividuals in Papua New Guinea showed erlands and Adegnika, Lambaréné, human therapies. Perhaps the realization
that infected individuals tended toward Gabon). Similarly, responses to vaccines that the same regulatory pathways that
higher IgE production although they against Salmonella were suppressed by are induced by helminth infections can
did not develop overt allergies (Daniel preexisting intestinal helminth infection also limit vaccine efficacy and interfere
Blount, Nottingham, England, UK). (Cathryn Nagler, Chicago, IL). Subop- with immunity to heterologous infec-
The practical potential of helminth in- timal responses in orally vaccinated mice tions will inject new energy into the ef-
fections in alleviating allergies and auto- were not caused by impaired antigen fort to use antihelmintic treatments to
immunity is not yet clear. The recognition, as activated, antigen-spe- maximize protection against the major
Nottingham group has completed phase I cific CD4+ T cells accumulated nor- childhood microbial diseases.
studies showing that low-dose hook- mally in the draining lymph nodes, but The authors are grateful for the generous support
worm infections are safe in humans and rather by increased antigen-specific, provided by the Keystone Symposia Global Health
do not exacerbate symptoms in rhinitis IL-10–producing CD4+ T cells—most Series, funded by the Bill and Melinda Gates
patients during hay fever season (Blount likely T reg cells. As one of the main Foundation. We are also grateful to the Burroughs
Wellcome Fund, the United European Gastroenterology
et al., 2009; Feary et al., 2009). Trials goals of global health organizations is to Federation, Cell Press, SCYNEXIS, and the National
are now under way to assess the poten- broaden vaccine coverage with a focus Institute of Allergy and Infectious Diseases for
tial benefits of this treatment in patients on geographical regions where gastroin- additional financial support for this meeting.

2064 immunoregulatory helminths | Maizels et al.


Meeting Review

REFERENCES King, S.B., A.M. Knorn, C. Ohnmacht, and D. sophil-CD4+ T cell interactions promote
Bazzone, L.E., P.M. Smith, L.I. Rutitzky, M.G. Voehringer. 2008. Accumulation of effector T(H)2 cytokine-dependent immunity. Nat.
Shainheit, J.F. Urban, T. Setiawan, A.M. CD4 T cells during type 2 immune responses Immunol. 10:697–705. doi:10.1038/ni.1740
Blum, J.V. Weinstock, and M.J. Stadecker. is negatively regulated by Stat6. J. Immunol. Pesce, J.T., T.R. Ramalingam, M.M. Mentink-
2008. Coinfection with the intestinal nema- 180:754–763. Kane, M.S. Wilson, K.C. El Kasmi, A.M.
tode Heligmosomoides polygyrus markedly Loke, P., I. Gallagher, M.G. Nair, X. Zang, F. Smith, R.W. Thompson, A.W. Cheever, P.J.
reduces hepatic egg-induced immunopathol- Brombacher, M. Mohrs, J.P. Allison, and Murray, and T.A. Wynn. 2009a. Arginase-1-
ogy and proinflammatory cytokines in mouse J.E. Allen. 2007. Alternative activation is an expressing macrophages suppress Th2 cyto-
models of severe schistosomiasis. Infect. Immun. innate response to injury that requires CD4+ kine-driven inflammation and fibrosis. PLoS
76:5164–5172. doi:10.1128/IAI.00673-08 T cells to be sustained during chronic infec- Pathog. 5:e1000371. doi:10.1371/journal.
Blount, D., D. Hooi, J. Feary, A. Venn, G. tion. J. Immunol. 179:3926–3936. ppat.1000371
Telford, A. Brown, J. Britton, and D. Loukas, A., J. Bethony, S. Brooker, and P. Hotez. Pesce, J.T., T.R. Ramalingam, M.S. Wilson,
Pritchard. 2009. Immunological profiles of 2006. Hookworm vaccines: past, present, M.M. Mentink-Kane, R.W. Thompson,
persons recruited for a randomized, placebo- and future. Lancet Infect. Dis. 6:733–741. A.W. Cheever, J.F. Urban Jr., and T.A.
controlled clinical trial of hookworm infec- doi:10.1016/S1473-3099(06)70630-2 Wynn. 2009b. Retnla (relmalpha/fizz1)
tion. Am. J. Trop. Med. Hyg. In press. Maizels, R.M. 2005. Infections and allergy - suppresses helminth-induced Th2-type
Butterworth, A.E., D.W. Dunne, A.J. Fulford, K.J. helminths, hygiene and host immune regu- immunity. PLoS Pathog. 5:e1000393.
Thorne, K. Gachuhi, J.H. Ouma, and R.F. lation. Curr. Opin. Immunol. 17:656–661. doi:10.1371/journal.ppat.1000393
Sturrock. 1992. Human immunity to Schistosoma doi:10.1016/j.coi.2005.09.001 Ramalingam, T.R., J.T. Pesce, M.M. Mentink-
mansoni: observations on mechanisms, and impli- Maizels, R.M., and M. Yazdanbakhsh. 2003. Kane, S. Madala, A.W. Cheever, M.R.
cations for control. Immunol. Invest. 21:391–407. Immune regulation by helminth parasites: Comeau, S.F. Ziegler, and T.A. Wynn.
doi:10.3109/08820139209069381 cellular and molecular mechanisms. Nat. Rev. 2009. Regulation of helminth-induced
Croese, J., J. O’neil, J. Masson, S. Cooke, W. Immunol. 3:733–743. doi:10.1038/nri1183 Th2 responses by thymic stromal lym-
Melrose, D. Pritchard, and R. Speare. Massacand, J.C., R.C. Stettler, R. Meier, N.E. phopoietin. J. Immunol. 182:6452–6459.
2006. A proof of concept study establish- Humphreys, R.K. Grencis, B.J. Marsland, and doi:10.4049/jimmunol.0900181
ing Necator americanus in Crohn’s patients N.L. Harris. 2009. Helminth products bypass Reinhardt, R.L., H.E. Liang, and R.M. Locksley.
and reservoir donors. Gut. 55:136–137. the need for TSLP in Th2 immune responses 2009. Cytokine-secreting follicular T cells
doi:10.1136/gut.2005.079129 by directly modulating dendritic cell func- shape the antibody repertoire. Nat. Immunol.
Elliott, D.E., A. Metwali, J. Leung, T. Setiawan, tion. Proc. Natl. Acad. Sci. USA. 106:13968– 10:385–393. doi:10.1038/ni.1715
A.M. Blum, M.N. Ince, L.E. Bazzone, M.J. 13973. doi:10.1073/pnas.0906367106 Rutitzky, L.I., L. Bazzone, M.G. Shainheit, B.
Stadecker, J.F. Urban Jr., and J.V. Weinstock. Metenou, S., B. Dembélé, S. Konate, H. Dolo, Joyce-Shaikh, D.J. Cua, and M.J. Stadecker.
2008. Colonization with Heligmosomoides S.Y. Coulibaly, Y.I. Coulibaly, A.A. Diallo, 2008. IL-23 is required for the development
polygyrus suppresses mucosal IL-17 produc- L. Soumaoro, M.E. Coulibaly, D. Sanogo, of severe egg-induced immunopathology in
tion. J. Immunol. 181:2414–2419. et al. 2009. Patent filarial infection modulates schistosomiasis and for lesional expression of
Everts, B., G. Perona-Wright, H.H. Smits, C.H. malaria-specific type 1 cytokine responses in an IL-17. J. Immunol. 180:2486–2495.
Hokke, A.J. van der Ham, C.M. Fitzsimmons, IL-10-dependent manner in a filaria/malaria- Shainheit, M.G., P.M. Smith, L.E. Bazzone, A.C.
M.J. Doenhoff, J. van der Bosch, K. Mohrs, coinfected population. J. Immunol. 183:916–924. Wang, L.I. Rutitzky, and M.J. Stadecker.
H. Haas, et al. 2009. Omega-1, a glycoprotein doi:10.4049/jimmunol.0900257 2008. Dendritic cell IL-23 and IL-1 production
secreted by Schistosoma mansoni eggs, drives Mwinzi, P.N., L. Ganley-Leal, C.L. Black, W.E. in response to schistosome eggs induces Th17
Th2 responses. J. Exp. Med. 206:1673–1680. Secor, D.M. Karanja, and D.G. Colley. cells in a mouse strain prone to severe immuno­
doi:10.1084/jem.20082460 2009. Circulating CD23+ B cell subset cor- pathology. J. Immunol. 181:8559–8567.
Feary, J., A. Venn, A. Brown, D. Hooi, F.H. relates with the development of resistance to Steinfelder, S., J.F. Andersen, J.L. Cannons, C.G.
Falcone, K. Mortimer, D.I. Pritchard, and J. Schistosoma mansoni reinfection in occupation- Feng, M. Joshi, D. Dwyer, P. Caspar, P.L.
Britton. 2009. Safety of hookworm infection ally exposed adults who have undergone mul- Schwartzberg, A. Sher, and D. Jankovic.
in individuals with measurable airway respon- tiple treatments. J. Infect. Dis. 199:272–279. 2009. The major component in schistosome
siveness: a randomized placebo-controlled fea- doi:10.1086/595792 eggs responsible for conditioning dendritic
sibility study. Clin. Exp. Allergy. 39:1060–1068. Mylonas, K.J., M.G. Nair, L. Prieto-Lafuente, cells for Th2 polarization is a T2 ribonucle-
doi:10.1111/j.1365-2222.2009.03187.x D. Paape, and J.E. Allen. 2009. Alternatively ase (omega-1). J. Exp. Med. 206:1681–1690.
Harnett, W., and M.M. Harnett. 2008. activated macrophages elicited by helminth doi:10.1084/jem.20082462
Therapeutic immunomodulators from nema- infection can be reprogrammed to enable mi- Taylor, M.D., N. van der Werf, A. Harris, A.L.
tode parasites. Expert Rev. Mol. Med. 10:e18. crobial killing. J. Immunol. 182:3084–3094. Graham, O. Bain, J.E. Allen, and R.M.
doi:10.1017/S1462399408000720 doi:10.4049/jimmunol.0803463 Maizels. 2009. Early recruitment of natu-
Hotez, P.J., P.J. Brindley, J.M. Bethony, C.H. Nair, M.G., Y. Du, J.G. Perrigoue, C. Zaph, ral CD4+ Foxp3+ Treg cells by infective
King, E.J. Pearce, and J. Jacobson. 2008. J.J. Taylor, M. Goldschmidt, G.P. Swain, larvae determines the outcome of filarial
Helminth infections: the great neglected trop- G.D. Yancopoulos, D.M. Valenzuela, A. infection. Eur. J. Immunol. 39:192–206.
ical diseases. J. Clin. Invest. 118:1311–1321. Murphy, et al. 2009. Alternatively acti- doi:10.1002/eji.200838727
doi:10.1172/JCI34261 vated macrophage-derived RELM-alpha Taylor, B.C., C. Zaph, A.E. Troy, Y. Du, K.J.
Hübner, M.P., J.T. Stocker, and E. Mitre. 2009. is a negative regulator of type 2 inflamma- Guild, M.R. Comeau, and D. Artis. 2009a.
Inhibition of type 1 diabetes in filaria-infected tion in the lung. J. Exp. Med. 206:937–952. TSLP regulates intestinal immunity and in-
non-obese diabetic mice is associated with a T doi:10.1084/jem.20082048 flammation in mouse models of helminth in-
helper type 2 shift and induction of FoxP3+ Ohnmacht, C., and D. Voehringer. 2009. Basophil fection and colitis. J. Exp. Med. 206:655–667.
regulatory T cells. Immunology. 127:512–522. effector function and homeostasis during doi:10.1084/jem.20081499
doi:10.1111/j.1365-2567.2008.02958.x helminth infection. Blood. 113:2816–2825. Taylor, J.J., C.M. Krawczyk, M. Mohrs, and
King, I.L., and M. Mohrs. 2009. IL-4–produc- doi:10.1182/blood-2008-05-154773 E.J. Pearce. 2009b. Th2 cell hyporespon-
ing CD4+ T cells in reactive lymph nodes Perrigoue, J.G., S.A. Saenz, M.C. Siracusa, E.J. siveness during chronic murine schistoso-
during helminth infection are T follicular Allenspach, B.C. Taylor, P.R. Giacomin, miasis is cell intrinsic and linked to GRAIL
helper cells. J. Exp. Med. 206:1001–1007. M.G. Nair, Y. Du, C. Zaph, N. van Rooijen, expression. J. Clin. Invest. 119:1019–1028.
doi:10.1084/jem.20090313 et al. 2009. MHC class II-dependent ba- doi:10.1172/JCI36534

JEM VOL. 206, September 28, 2009 2065


Thompson, R.W., J.T. Pesce, T. Ramalingam, flammation by helminth-induced regula- stasis. Nature. 446:552–556. doi:10.1038/
M.S. Wilson, S. White, A.W. Cheever, S.M. tory T cells. J. Exp. Med. 202:1199–1212. nature05590
Ricklefs, S.F. Porcella, L. Li, L.G. Ellies, and doi:10.1084/jem.20042572 Zaph, C., Y. Du, S.A. Saenz, M.G. Nair, J.G.
T.A. Wynn. 2008. Cationic amino acid trans- Wynn, T.A. 2007. Common and unique mecha- Perrigoue, B.C. Taylor, A.E. Troy, D.E.
porter-2 regulates immunity by modulating nisms regulate fibrosis in various fibroprolif- Kobuley, R.A. Kastelein, D.J. Cua, et al.
arginase activity. PLoS Pathog. 4:e1000023. erative diseases. J. Clin. Invest. 117:524–529. 2008. Commensal-dependent expression
doi:10.1371/journal.ppat.1000023 doi:10.1172/JCI31487 of IL-25 regulates the IL-23-IL-17 axis in
van Panhuys, N., S.C. Tang, M. Prout, M. Wynn, T.A. 2008. Cellular and molecular mech- the intestine. J. Exp. Med. 205:2191–2198.
Camberis, D. Scarlett, J. Roberts, J. Hu-Li, anisms of fibrosis. J. Pathol. 214:199–210. doi:10.1084/jem.20080720
W.E. Paul, and G. Le Gros. 2008. In vivo doi:10.1002/path.2277 Zaretsky, A.G., J.J. Taylor, I.L. King, F.A.
studies fail to reveal a role for IL-4 or STAT6 Zaccone, P., O. Burton, N. Miller, F.M. Marshall, M. Mohrs, and E.J. Pearce.
signaling in Th2 lymphocyte differentiation. Jones, D.W. Dunne, and A. Cooke. 2009. 2009. T follicular helper cells differenti-
Proc. Natl. Acad. Sci. USA. 105:12423–12428. Schistosoma mansoni egg antigens induce Treg ate from Th2 cells in response to hel-
doi:10.1073/pnas.0806372105 that participate in diabetes prevention in minth antigens. J. Exp. Med. 206:991–999.
Wilson, M.S., and R.M. Maizels. 2004. Regulation NOD mice. Eur. J. Immunol. 39:1098–1107. doi:10.1084/jem.20090303
of allergy and autoimmunity in helminth in- doi:10.1002/eji.200838871 Zenewicz, L.A., G.D. Yancopoulos, D.M.
fection. Clin. Rev. Allergy Immunol. 26:35–50. Zaph, C., A.E. Troy, B.C. Taylor, L.D. Berman- Valenzuela, A.J. Murphy, S. Stevens, and
doi:10.1385/CRIAI:26:1:35 Booty, K.J. Guild, Y. Du, E.A. Yost, A.D. R.A. Flavell. 2008. Innate and adaptive in-
Wilson, M.S., M.D. Taylor, A. Balic, C.A.M. Gruber, M.J. May, F.R. Greten, et al. 2007. terleukin-22 protects mice from inflamma-
Finney, J.R. Lamb, and R.M. Maizels. Epithelial-cell-intrinsic IKK-beta expres- tory bowel disease. Immunity. 29:947–957.
2005. Suppression of allergic airway in- sion regulates intestinal immune homeo- doi:10.1016/j.immuni.2008.11.003

2066 immunoregulatory helminths | Maizels et al.

View publication stats

You might also like