4
Immunoparasitology Phillip Scott Author’s address Phillip Scott, Department of Pathobiology, School of Veter- inary Medicine, University of Pennsylvania, Philadelphia, PA, USA. Correspondence to: Phillip Scott Department of Pathobiology School of Veterinary Medicine 3800 Spruce Street Philadelphia, PA 19104, USA Fax: þ1 215 573 7023 E-mail: [email protected] Mosmann et al. (1) reported that CD4 þ T-cell clones could be divided into two subsets based on the cytokines they pro- duced. Understanding these subsets, termed T-helper 1 (Th1) and Th2, and more broadly defining the role of cyto- kines in protection and immunopathology have been the basis for much of the research in the field of immunoparasitology over the last 18 years. Murine models of parasitic diseases, such as leishmaniasis and trichuriasis, provided the oppor- tunity to determine how Th1 and Th2 cells differentiate and regulate each other in an in vivo context. Studies with both protozoan parasites and tissue-dwelling helminths helped elucidate the cells and cytokines that shape the adaptive immune response, and studies with gastrointestinal nematodes explored the poorly understood immune system of the gut. In this volume, many of those contributions are highlighted. Taken together, they point out how the field of immuno- parasitology has contributed to our understanding of the immune response. While it is hoped that this information will lead to better treatments and vaccines for parasitic diseases, on this front there has been only limited success; there are no vaccines for any of the major human parasitic diseases. However, several contributions in this volume address this issue and describe how immunologic memory functions in parasitic diseases – information that will be useful for developing successful vaccines. After the initial discovery that interleukin-12 (IL-12) and IL-4 promoted Th1 and Th2 responses respectively, a series of questions were raised (2). How and when are these cytokines produced after infection? What cells produce IL-12 and IL-4, and where are these cells located? What parasite molecules activate cells to produce these cytokines? What interactions with the host lead to preferential development of Th1 responses after some infections (e.g. Toxoplasma) and Th2 responses after others (e.g. many of the helminths), and why are some parasites able to induce both responses – either at different stages of the infection (e.g. Schistosoma mansoni , malaria) or Immunological Reviews 2004 Vol. 201: 5–8 Printed in Denmark. All rights reserved Copyright ß Blackwell Munksgaard 2004 Immunological Reviews 0105-2896 5

Immunoparasitology

Embed Size (px)

Citation preview

ImmunoparasitologyPhillip Scott

Author’s address

Phillip Scott,Department of Pathobiology, School of Veter-

inary Medicine, University of Pennsylvania,

Philadelphia, PA, USA.

Correspondence to:

Phillip ScottDepartment of Pathobiology

School of Veterinary Medicine

3800 Spruce Street

Philadelphia, PA 19104, USA

Fax: þ1 215 573 7023

E-mail: [email protected]

Mosmann et al. (1) reported that CD4þ T-cell clones could be

divided into two subsets based on the cytokines they pro-

duced. Understanding these subsets, termed T-helper 1

(Th1) and Th2, and more broadly defining the role of cyto-

kines in protection and immunopathology have been the basis

for much of the research in the field of immunoparasitology

over the last 18 years. Murine models of parasitic diseases,

such as leishmaniasis and trichuriasis, provided the oppor-

tunity to determine how Th1 and Th2 cells differentiate and

regulate each other in an in vivo context. Studies with both

protozoan parasites and tissue-dwelling helminths helped

elucidate the cells and cytokines that shape the adaptive

immune response, and studies with gastrointestinal nematodes

explored the poorly understood immune system of the gut. In

this volume, many of those contributions are highlighted.

Taken together, they point out how the field of immuno-

parasitology has contributed to our understanding of the

immune response. While it is hoped that this information

will lead to better treatments and vaccines for parasitic

diseases, on this front there has been only limited success;

there are no vaccines for any of the major human parasitic

diseases. However, several contributions in this volume

address this issue and describe how immunologic memory

functions in parasitic diseases – information that will be useful

for developing successful vaccines.

After the initial discovery that interleukin-12 (IL-12) and

IL-4 promoted Th1 and Th2 responses respectively, a series of

questions were raised (2). How and when are these cytokines

produced after infection? What cells produce IL-12 and IL-4,

and where are these cells located? What parasite molecules

activate cells to produce these cytokines? What interactions

with the host lead to preferential development of Th1

responses after some infections (e.g. Toxoplasma) and Th2

responses after others (e.g. many of the helminths), and why

are some parasites able to induce both responses – either at

different stages of the infection (e.g. Schistosoma mansoni, malaria) or

Immunological Reviews 2004Vol. 201: 5–8Printed in Denmark. All rights reserved

Copyright � Blackwell Munksgaard 2004

Immunological Reviews0105-2896

5

in different inbred strains of mice (e.g. Leishmania, Trichuris)? What

are the effector mechanisms involved in protection against

gastrointestinal nematodes? Some of these questions remain

unanswered, but the focus of the last several years on early

host–pathogen interactions has provided information that has

significantly increased our understanding in this area.

In this volume, several laboratories report on the interac-

tions between protozoa and the signaling pathways associated

with innate immune responses. Protozoa promote a Th1

response through interactions with the innate immune system,

presumably utilizing signaling pathways employed by other

pathogens. Gazzinelli, Sher, Langhorne, and their colleagues

describe the parasite molecules from Trypanosoma cruzi, African

trypanosomiasis, Toxoplasma gondii, Leishmania, and malaria that

can promote the activation of the innate immune system.

Many of these molecules contain glycosylphosphatidylinosital

anchors that appear to function as pathogen-associated mole-

cular patterns (PAMPs) and thus activate Toll/IL-1 receptor-

signaling pathways, although for several of these molecules, it

has been difficult to assign a specific Toll-like receptor (TLR)

to their function. In addition to the role that TLRs play in

parasite activation of cells, other pathways also contribute to

cell activation. For example, in Toxoplasma, a parasite cyclophi-

lin was described that binds to CCR5 and induces IL-12 pro-

duction. Because the nuclear factor-k B (NF-kB) family of

transcription factors is central to the development of both

innate and adaptive immune responses, it is important to

define how these different NF-kB transcription factors func-

tion in parasitic diseases. In this volume, Hunter and cowork-

ers review what is known about NF-kB in the development of

immunity to Toxoplasma, Leishmania, and Trichuris.

The factors contributing to the development of Th2

responses following infection with helminths are less well

defined. In this volume, contributions from the laboratories

of Gause, Grencis, and Maizels address this issue. Owing to the

association of TLRs and Th1 responses, it may not be surpris-

ing that no parasite TLR ligands capable of stimulating Th2

responses have been defined. However, that does not mean

that helminths do not express TLR ligands. Thus, Trichuris muris

stimulates the Toll pathway, but rather than initiating a pro-

tective response, such activation is associated with suscept-

ibility. This leaves open the question of how helminths

provoke such a dominant Th2 response. Some suggest that

the preferential development of Th2 responses is associated

with the release of helminth proteases (Maizels et al.), while

others report that helminth antigens that have no proteolytic

activity can still promote Th2 responses (Gause et al.). Clearly,

there are likely to be multiple pathways involved, and the

studies reported here provide an extensive overview of this

field.

One approach to defining the factors promoting T-helper

cell development is to study dendritic cells (DCs), as these cells

initiate immune responses. As described above, protozoa may

utilize Toll/IL-1 receptor-signaling pathways, as well as other

pathways, to stimulate DCs and promote a Th1 response. As

discussed by Langhorne and colleagues, the nature of the

interactions between DCs and the blood stages of malaria

will dictate the nature of the immune response that develops.

These observations with protozoan parasites raise the question

of how helminths influence DC function and whether hel-

minths actively promote a Th2 response by stimulating DCs

in a particular way. One of the most studied parasitic hel-

minths is Schistosoma mansoni, in which the role of DCs has been

extensively investigated. Schistosomiasis is associated with an

early Th1 response that switches to a dominant Th2 response

over time. This switch occurs when egg laying begins, and in

this volume, Pearce et al. describe how egg antigens influence

DC function, such that they promote Th2 responses. Import-

antly, it appears that once DCs are exposed to egg antigens,

they can promote a Th2 response by themselves, highlighting

the critical role of DCs in shaping the nature of the immune

response in this infection. Nutman and colleagues, in their

studies with filariasis, further highlight the complexity of

parasite–DC interactions. The complex filarial life cycle leads

to the presence of different parasite stages interacting with DCs

in different locations and potentially leading to different out-

comes. Notably, live microfilaria were found to induce DC

apoptosis, although the mechanism involved is not yet

defined, while exposure of human Langerhans’ cells to third

stage larvae leads to DC migration but not upregulation of

markers associated with activation.

One of the unique areas of research in the field of immuno-

parasitology is the identification of effector pathways asso-

ciated with protection to gastrointestinal helminths, and the

contributions by Finkelman, Grencis, and Maizels discuss these

pathways. When one considers that there are more than 3.5

billion people infected with these organisms, it is astonishing

that the final effector mechanism(s) involved in controlling

these parasites is still poorly understood. Nevertheless, there is

a consensus emerging that while infection with gut nematodes

induces many effector mechanisms, only some of them are

effective against a particular species. The effector mechanisms

involved in controlling tissue-dwelling helminths are also an

area of controversy; in this volume, Wynn, Stadecker, Dessein,

and their coworkers address the relative contributions of

the Th1 and Th2 immune response to protection and

Scott � Immunoparasitology

6 Immunological Reviews 201/2004

immunopathology associated with schistosomiasis. What is

clear is that an appropriate balance in the Th1/Th2 ratio

is critical in controlling immunopathology, and how that is

achieved is an area of intensive investigation.

The importance of regulating host effector mechanisms may

stem from the fact that in contrast to many bacterial and viral

infections, most parasitic diseases are chronic, thus putting

pressure on the host to modulate effector responses that if

continued would be immunopathologic. In fact, parasitic dis-

ease is frequently not the result of the absence of immunity

but rather is due to the continued presence of an ineffective

immune response. This immunopathology may be a disadvan-

tage not only for the host but also for the parasite; thus, there

are many strategies employed by parasites to modulate

immune effector mechanisms. Some occur quite early after

infection. For example, in this volume, Denkers et al. describe

how Toxoplasma blocks both the NF-kB and the mitogen-

activated protein kinase pathways – critical signaling pathways

promoting proinflammatory responses. Interestingly, Toxo-

plasma may also block the CCR5-dependent pathway for IL-12

production (described above) by releasing a parasite product

related to lipoxin, an arachidonic acid metabolite that down-

regulates CCR5.

As Th1 and Th2 cells cross-regulate each other via suppres-

sive cytokines, much of the focus on regulation has been on

these cells and the cross-regulatory cytokines they produce.

However, many other regulatory mechanisms exist. For exam-

ple, as described by Wynn et al. in this volume, a decoy IL-13

receptor is critical for controlling the egg-induced granuloma-

tous response associated with S. mansoni. Furthermore, chronic

disease may not always be the result of cross-regulation by

Th1 and Th2 cells. Thus, in this volume, McMahon-Pratt and

Alexander describe the immune responses associated with the

chronic disease observed following infection with L. mexicana

and L. donovani parasites. In contrast to L. major infections, the

inability to resolve an infection with these organisms is not

due to a dominant Th2 response, suggesting that other

mechanisms suppress the development of protective Th1

responses. How this suppression occurs is actively being pur-

sued by several laboratories.

A particularly exciting area of immunoregulation is defining

the role of T-regulatory (Treg) cells in modulating the

immune responses to parasites. Treg cells were initially

described as cells that control autoimmunity, but we now

know that they also play a critical role in modulating immune

responses induced by infection (3). In this volume, an article

from the laboratory of Sacks describes the important role Treg

cells play in leishmaniasis. Interestingly, it appears that the

presence of Treg cells is required to maintain persistent para-

sites after resolution of disease. This finding is of particular

note, because in the absence of persistent parasites, a large part

of the immunity associated with resolving a primary infection

with Leishmania was lost. While the initial focus has been on the

ability of Treg cell to suppress Th1 responses, Maizels and

colleagues describe the potential role of Treg cells in modulat-

ing Th2 responses associated with helminth infections. Thus,

in a mouse model of filariasis, depletion of Treg cells led to

heightened Th2 effector activity and clearance of a large per-

centage of the worms. These results raise the obvious next

question of how Treg cells are activated during infection;

levels of Treg cells do appear to fluctuate during infection,

but how this occurs is unknown. An interesting observation

reported by Kaye et al. is that stromal cells taken from L.

donovani-infected mice promote the development of DCs that

may contribute to Treg-cell development or expansion. Many

unanswered questions about these cells exist, and studies in

models of parasitic diseases are likely to help answer some of

these questions.

Despite the significant advances we have made in our under-

standing of the immune responses that occur following para-

sitic infections, no vaccine for any of these major human

diseases is currently in use. The difficulties in the development

of a successful vaccine include defining protective antigens,

determining what is the most appropriate effector mechanism

to elicit, determining what adjuvants to use, and understand-

ing the basic immunology associated with memory-cell devel-

opment and maintenance. The development of a vaccine for

human malaria, which causes more than two million deaths a

year, has been a high priority for many years. Nevertheless, no

vaccine has been forthcoming. In this issue, Good et al.

describe the challenges associated with the development of a

malaria vaccine and suggest that a successful approach may not

depend on immunity that mimics natural immunity. How-

ever, this discussion does not mean that after natural infec-

tions, malaria is unable to induce some level of immunity, and

in this volume, Riley and colleagues provide an important

analysis of the memory responses that are associated with

malaria infection. Understanding the limitations of naturally

induced memory will be critical in forging a new approach to

vaccine development.

The final three articles focus on understanding how early

effector responses and subsequent memory responses develop

during parasitic infections. The development of efficacious

vaccines remains a challenge for the field of parasitology,

and one of the problems may be that so little is understood

about the generation of memory cells, particularly memory

Scott � Immunoparasitology

Immunological Reviews 201/2004 7

T cells. Recently, several laboratories have characterized the

memory T cells arising following bacterial or viral infections,

which has provided a better understanding of memory T cells

(4). These studies and others indicate that subpopulations of

memory T cells exhibit distinct functions and migration pat-

terns. However, as parasitic infections are frequently chronic,

the factors involved in the development and maintenance of

memory T cells may differ from acute bacterial or viral infec-

tions where sterile immunity is achieved. In this volume,

articles from the laboratories of Zavala and Tarleton define

the generation and maintenance of effector and memory

CD8þ T cells during infection with Plasmodium and T. cruzi,

respectively. Both of these studies utilize T-cell receptor trans-

genic mice to monitor the in vivo activation and expansion of T

cells after infection, permitting a sophisticated analysis of the

factors involved in both T-cell effector function and memory

T-cell development. For example, in T. cruzi, the capacity of

CD8þ T cells to provide protection was completely dependent

upon their ability to make interferon-g, and in malaria the

ability of CD4þ T cells to promote CD8þ T-cell responses was

shown to depend upon IL-4 production. Finally, we describe

how CD4þ Th1 cells develop and are maintained as memory

CD4þ T cells during infection with Leishmania major. These

studies indicate that infection-induced resistance to L. major is

mediated by both an effector T-cell population, maintained

due to the persistence of parasites following resolution of

disease, and a central memory T-cell population that does

not require persistent parasites.

The work described in this volume provides an up-to-date

picture of where the field is at present; from these studies, one

can predict what the future directions are for immunopara-

sitology. Clearly much more information on the PAMPs and

other parasite factors that induce both Th1 and Th2 responses

is required; this information may aid in designing immuno-

therapies. Gastrointestinal nematodes remain a huge burden

on humankind, and better understanding of mucosal immune

responses will be a focus of much research. Regulation of

immunity is a hallmark of parasitic infection, and future stud-

ies are likely to focus on a better understanding of how Treg

cells develop and function in a number of parasitic diseases.

Finally, we ended this volume with chapters addressing how

memory T cells develop; this area will also be one of continued

extensive research, which hopefully will not only broaden our

understanding of immunologic memory but also provide

information useful in the design of the much needed vaccines

for parasites.

References

1. Mosmann TR, Cherwinski H, Bond MW,

Giedlin MA, Coffman RL. Two types of

murine helper T cell clone. I. Definition

according to profiles of lymphokine activities

and secreted proteins. J Immunol

1986;136:2348–2357.

2. Murphy KM, Reiner SL. The lineage decisions

of helper T cells. Nat Rev Immunol

2002;2:933–944.

3. Hori S, Takahashi T, Sakaguchi S. Control of

autoimmunity by naturally arising regulatory

CD4þ T cells. Adv Immunol 2003;81:331–371.

4. Sallusto F, Geginat J, Lanzavecchia A. Central

memory and effector memory T cell subsets:

function, generation, and maintenance. Annu

Rev Immunol 2004;22:745–763.

Scott � Immunoparasitology

8 Immunological Reviews 201/2004