You are on page 1of 7

272 Review TRENDS in Parasitology Vol.18 No.

6 June 2002

39 Urdaneta, L. et al. (2001) Evidence for clonal 41 Jongwutiwes, S. et al. (1994) Allelic variation in Acad. Sci. U. S. A. 97, 6994–7001
propagation in natural isolates of Plasmodium the circumsporozoite protein of Plasmodium 43 Winzeler, E.A. et al. (1998) Direct allelic variation
falciparum from Venezuela. Proc. Natl. Acad. Sci. falciparum from Thai field isolates. Am. J. Trop. scanning of the yeast genome. Science 281,
U. S. A. 98, 6725–6729 Med. Hyg. 51, 659–668 1194–1197
40 Maynard Smith, J. and Haigh, J. (1974) The 42 Rich, S.M. and Ayala, F.J. (2000) 44 Desowitz, R.S. (1991) The Malaria Capers: More
hitch-hiking effect of a favorable gene. Genet. Res. Population structure and recent evolution Tales of Parasites and People, Research and
23, 23–35 of Plasmodium falciparum. Proc. Natl. Reality, W.W. Norton

How protozoan parasites evade the


immune response
Sergio Zambrano-Villa, Disney Rosales-Borjas, Julio César Carrero and
Librado Ortiz-Ortiz

Protozoan pathogens such as Plasmodium, Leishmania, Trypanosoma and stages. During the very early stage of infection, the
Entamoeba are responsible for several of the most widespread and lethal host response is poor because of the relatively low
human diseases. Their successful survival depends mainly on evading the host density of sporozoites and their rapid migration to
immune system by, for example, penetrating and multiplying within cells, the liver. In order to survive, the parasite must reach
varying their surface antigens, eliminating their protein coat, and modulating the hepatocytes and transform into merozoites,
the host immune response. Immunosuppression is sometimes caused directly acquiring new biological features to confront the
by parasite products and sometimes involves antigenic mimicry, which often immune system. Once the Plasmodium parasite is
appears in association with parasitic diseases. However, one of the most established in the host, it evades the immune
sophisticated mechanisms of evasion is the selective activation of a subset response (IR) by changing its surface antigens
of T helper cells. as it passes through the stages of its life cycle.
Thus, each phase of the cellular cycle is associated
Parasites that cause malaria, sleeping sickness, with the expression of stage- and species-specific
Chagas disease, amoebiasis and leishmaniasis proteins, many of which are inserted into the
primarily affect the populations of developing membrane surface of the parasite or the red blood
countries, causing high rates of morbidity and cells they invade.
mortality [1]. The host’s mechanisms of defense range The development of an effective IR is hampered by
from primary barriers to the most elaborate devices, the fact that stage-specific proteins tend to be highly
which involve a wide variety of cells and molecules polymorphic or antigenically variable. Sequence
capable of the specific recognition and elimination of polymorphism is common in several Plasmodium
invasive agents. Parasites with extracellular stages proteins, especially in those with extensive repetitive
are the target of the humoral immune response, regions, such as the circumsporozoite protein (CSP),
whereas those with intracellular stages are an antigen with multiple tandem repeats and that is
Sergio Zambrano-Villa
Centro Universitario de susceptible to attack by the cell-mediated immunity. located on the surface of malaria sporozoites.
Ciencias Exactas e Despite the large amount of antigen presented by the Antigens with multiple repeats could downregulate
Ingenierías, Universidad parasite to the host and the host’s immune and antibody isotype maturation and the production of
de Guadalajara,
Guadalajara, Jalisco,
inflammatory response, parasites manage to survive high-affinity antibodies by acting as B-cell
México. within the host for long periods by using multiple superantigens and predominantly inducing a
evasion mechanisms that have been acquired during polyclonal thymus-independent humoral response
Disney Rosales-Borjas
Hospital Universitario Dr millions of years of evolution (Table 1). Parasites have [2]. Despite the short time of circulation of the
Miguel Oraá, Guanare, evasion mechanisms that depend on factors such as malaria extracellular phases, when an antibody
Edo. Portuguesa, their life-cycle stage, the route of penetration and the response does develop against sporozoites,
Venezuela.
microenvironment in which they are established plasmodia overcome the response by sloughing off
Julio César Carrero inside the host. their surface CSP coat, rendering the antibodies
Librado Ortiz-Ortiz*
Departamento de
ineffective. The versatility of CSP is also evident in its
Inmunología, Instituto de Malaria RNA-binding motifs, which block RNA translation in
Investigaciones In humans, malaria is caused by Plasmodium vivax, mammalian cells [3].
Biomédicas, UNAM, Plasmodium malariae, Plasmodium ovale and Other proteins, such as the P. falciparum
México.
*e-mail: librado@
Plasmodium falciparum. The infection involves a merozoite surface protein (MSP-1) 1, induce ‘blocking
servidor.unam.mx complex life cycle with intra- and extracellular antibodies’ that bind anywhere on MSP-1, preventing

http://parasites.trends.com 1471-4922/02/$ – see front matter © 2002 Elsevier Science Ltd. All rights reserved. PII: S1471-4922(02)02289-4
Review TRENDS in Parasitology Vol.18 No.6 June 2002 273

Table 1. Comparison of main evasion mechanisms for selected protozoan parasitesa


Parasite (disease) Epidemiology of the Main strategies of evasion Result Refs
disease [1]
Plasmodium falciparum 300 million–500 million Antigenic variation and/or polymorphisms Evades the IR; infected cells are [2]
(malaria) infected per year prevented from being swept to the
1.5 million–2.7 million spleen
deaths per year Induction of blocking antibodies Blocks the binding of real inhibitory [4]
antibodies
Molecular mimicry Alters immune recognition and might [6,7]
induce autoimmune disease
Anergy of T cells Immunosuppression [10–14]
Altered peptide ligand Alters functions of memory T cells [15,16]
Trypanosoma brucei 15 000–20 000 new cases Antigenic variation by VSG Evades the previously established IR [17,18]
(African trypanosomiasis per year Alteration of T- and B-cell populations Immunosuppression [19]
or sleeping sickness) 55 million at risk Abnormal activation of macrophages Impairs macrophage functions [20]
Induction of changes in the pattern of cytokines Increases IFN-γ and decreases IL-2 and [21]
released by CD8+ T cells IL-2R; renders T cells unresponsive
Production of a gp63-like-protein Resists complement [22]
Trypanosoma cruzi 12 million–16 million Increased phagocytic activity More CD8+ T cells and reduced TDR [23]
(Chagas disease) infected and TIR
45 000 deaths per year Parasite mucin that binds to macrophage Impairs macrophage cytokine secretion [25]
90 million at risk Anergy of T cells Immunosuppression [26,27]
Production of blocking IgM antibodies Blocks the binding of real inhibitory [29]
antibodies
Turnover of surface molecules, phospholipases Resists complement [31–33]
and complement-regulating factors
Entamoeba histolytica 500 million infected Cytolytic capacity Damages host cells and tissues,
(Intestinal and liver 40 000–110 000 deaths interfering with IR
amoebiasis) per year Degradation of antibodies by proteases Evades humoral immunity [34]
Acquisition of complement-regulating factors, Resists complement and protects [36,37]
shedding of immune complexes by capping against the inflammatory response
and inactivation of C3a and C5a [38,39]
Anergy of T cells Immunosuppression [41–43]
Release of products (MLIF among others) that Impairs macrophage functions
act on macrophages; produces PGE2
Induction of IL-4 and IL-10 Modulates the Th1 response [45]
Leishmania parasites 12 million infected Inhibition of phagolysosome formation and Evades the macrophage proteolytic [46,47]
(leishmaniasis: 350 million at risk the proteolytic enzymes from lysosome processes
cutaneous, 1 million–1.5 million new [48]
mucocutaneous and cases per year of Abnormal activation of protein kinase C and Inhibits the respiratory burst [49]
visceral) cutaneous leishmaniasis scavenging of oxygen intermediates
500 000 new cases and Prevention of apoptosis of infected Extends the survival of the infected [50,51,
75 000–80 000 deaths macrophage macrophage 53]
per year owing to Inhibition of MHC-protein production, peptide Impairs macrophage antigen- [54,56]
visceral leishmaniasis loading and expression of co-stimulatory presenting function [57]
molecules
[58,59]
Induction of PGE2 and downregulation of TNF-αR Impairs macrophage function
Inhibition of macrophage and neutrophil Promotes survival of parasites once [61]
chemotaxis established
Shedding of MAC complex and inactivation of Resists complement
some MAC components
Suppression of transcription of the IL-12 gene Blocks the protective Th1 response
aAbbreviations: IFN-γ, interferon γ; IL-2, interleukin 2; IL-4, interleukin 4; IL-10, interleukin 10; IL-12, interleukin 12; IL-2R, IL-2 receptor; IR, immune response; MAC, membrane
attack complex; MHC, major histocompatibility complex; MLIF, monocyte locomotion inhibition factor; PGE2, prostaglandin E2; TDR, thymus-dependent response;
TIR, thymus-independent response; TNF-αR, tumor necrosis factor α receptor; VSG, variant surface glycoprotein.

the binding of antibodies with real inhibitory their significantly altered morphology. To circumvent
capacity [4]. In addition, the P. falciparum protein the effect of anti-PfEMP-1 antibodies, the molecule
PfEMP-1, which is expressed at the surface of infected undergoes clonal variation at high frequency [5].
erythrocytes, favors adherence to the vascular Molecular mimicry, which is common in human
endothelium via molecules such as CD36 and malaria, can modulate the cellular IR and the release
ICAM-1. In this way, the infected cells are prevented of cytokines and is probably involved in some of
from being swept to the spleen, where they would be the pathological manifestations. Thus, the
recognized as altered and phagocytosed because of glycosylphosphatidylinositol (GPI) moiety that

http://parasites.trends.com
274 Review TRENDS in Parasitology Vol.18 No.6 June 2002

anchors a range of MSPs appears to have a marked are closely related to those of the original CSP. These
insulin-mimetic effect, reducing the glucose levels in variants prevent the binding of the original T-cell
rodents at the same time that it increases the epitopes to the major histocompatibility complex
synthesis of tumor necrosis factor (TNF) α and (MHC) molecule, altering memory-T-cell effector
interleukin-1 (IL-1) in macrophages; this is functions, such as lymphokine synthesis, cytotoxicity
responsible for the typical malarial fevers and the and proliferation [15]. During the liver stage, CSP
production of acute-phase proteins [6]. In the same participates in the binding of sporozoites to
context, sequences homologous to 1-thymosin hepatocytes and contains cytotoxic T-lymphocyte
(a thymic hormone that modulates the differentiation (CTL) epitopes that bind common MHC-class-I
of T cells) have been identified in two P. falciparum alleles. A pair of CSP variant epitopes have been
proteins. Like the human hormone, synthetic peptides identified in parasites from The Gambia that did not
from those sequences showed a similar effect on T-cell bind to the MHC-class-I allele HLA-B35 or induce
maturation, suggesting that it might interfere with CTL responses [16], by contrast to the original epitopes.
the development of effective cellular immunity [7]. This is particularly important if we consider that CTL
Moreover, molecular mimicry in combination with the and the memory response appear to be essential for
polyclonal B-cell activation (PBCA) could be protection and long-lasting immunity, respectively.
responsible for the raising of autoantibodies in
human malaria. Autoantibodies have been found African trypanosomiasis
against single-stranded DNA, heat-shock protein 70 African trypanosomiasis (sleeping sickness) is
and polypeptide and carbohydrate epitopes from red produced in humans by two African trypanosomes
blood cells [8]. However, the association between (Trypanosoma brucei gambiense and T. brucei
autoantibodies and malaria pathology is rhodesiense) and in cattle by four (T. brucei brucei,
controversial. Moreover, it has been suggested that Trypanosoma vivax, Trypanosoma evansi and
autoantibodies could have a role in immune protection Trypanosoma congolense). These trypanosomes live in
against malaria and other infectious diseases [9]. the blood and lack intracellular stages, so the parasite
In comparison with these surface proteins, some is a target for antibody-mediated destruction.
internal antigens are less variable and seem to be at Certainly, almost all trypanosomes are cleared from
least partially conserved among Plasmodium species. blood by liver macrophages. However, the remaining
When the schizont bursts, some of the antigens parasites survive and establish the infection thanks
released in large quantities appear also to be involved to the antigenic variation of the so-called variant
in triggering the cascade of cytokines responsible for surface glycoprotein (VSG). Each trypanosome
most of the symptoms and pathology of the infection. carries a large repertoire of VSG genes coding for
Evidence suggests that acute malaria infection multiple VSG variants with different primary
induces a temporal reduction of the IR. During sequence, particularly at the N termini [17].
infection with P. falciparum, circulating T cells are Interestingly, each parasite expresses a single VSG
reduced in number, accompanied by a decrease in gene at any one time by replacing the previous gene at
lymphoproliferative response and cytokine release by the telomeric active site of transcriptional expression
peripheral blood mononuclear cells when stimulated with the new one [18]. In consequence, this repeated
by malaria antigens [10]. This effect is partially antigenic change of VSG in trypanosomes allows
reverted by the addition of indomethacin to the cell them to evade the TDHR, resulting in successive
cultures, indicating that prostaglandin secreted by surges of parasitemia, a situation similar to being
activated macrophages might be responsible [11]. infected successively by related, but not identical,
Alternatively, it might be caused by the release into pathogens. This phenomenon makes it difficult to
the serum of acute-phase proteins that bind to the develop a vaccine against the disease.
surface of lymphoid cells, inhibiting lymphocyte Because pathogenesis is linked to the inability of the
proliferation [12]. However, it has also been untreated patient to eliminate the parasite, attempts
reported that some malaria antigens can directly have been made to determine the way by which the
induce immunosuppression. A low molecular parasite interacts with the immune system and
weight glycoprotein has been isolated from disturbs the balance of cytokine and other mediators,
Plasmodium berghei that suppresses the primary thus allowing the pathology to progress. During
thymus-dependent humoral response (TDHR) to parasitemia, several populations of B and T cells
antigens in mice [13] and a schizont extract from are altered. The TDHR against the persistent
P. falciparum suppresses the lymphoproliferative trypanosome antigens is depressed, although the
response to malarial and other soluble antigens in vitro thymus-independent humoral response to the VSG
[14]. The mechanisms responsible for suppression are surface epitopes [19] can probably control subsequent
not well known, although it has been suggested that parasitemias. Trypanosome products also activate
parasite hemozoin accumulation in macrophages macrophages and CD8+ T cells in a particular way that
might inhibit their accessory function. Recent analysis causes changes in the pattern of cytokines they release.
of the IR against CSP suggests that some P. falciparum Among these, the GPI moiety of VSG appears to
strains contain CSP variants with T-cell epitopes that overactivate the macrophages, which become

http://parasites.trends.com
Review TRENDS in Parasitology Vol.18 No.6 June 2002 275

suppressor macrophages that produce TNF-α [20] and, T-cell immunosuppression during experimental
similar to the parasite protein T-cell-triggering factor, infection by T. cruzi has been confirmed by various
induce CD8+ (but not CD4+) T cells to secrete high levels authors. Parasite products interfere with various
of interferon (IFN) γ [21]. In turn, the high levels of IFN-γ components of the immune system, including a
result in the decrease of IL-2 receptor expression and decrease in the expression of lymphocyte receptors
IL-2 synthesis, which impair the proliferative T-cell (e.g. IL-2R) [27] and in the production of cytokines
response to avoid parasite elimination. involved in the effector phase of the IR [26]. The
Recently, a set of experiments have shown that mechanism is unknown but could involve suppressor
trypanosomes have several homologous genes cells characterized as spleen non-adherent Thy-1+
encoding glycoprotein 63 (gp63)-like proteins, similar Ly-2+ cells [28]. Trypanosoma cruzi, similar to other
to those from Leishmania [22]. Although their role in protozoa, induces PBCA with subsequent
trypanosomes has not yet been determined, the overproduction of IgM antibodies. These antibodies
correlation between the amounts of gp63-like-protein bind to the trypomastigote surface, interfering with
on the surface of different stages of the parasite and the binding of IgG inhibitory antibodies and
their susceptibility to complement-mediated lysis consequently preventing the elimination of T. cruzi
suggests an important role in immune evasion at the parasites in mice [29]. Moreover, PBCA could activate
blood stage. Thus, the resistance of blood B cells specific for autoantigens, potentially inducing
trypanosomes, which contain a lot of gp63-like- the autoimmune disease that is characteristic of this
protein, contrasts with the susceptibility of procyclic infection [30]. In the same context, T. cruzi produces
trypanosomes, which have less. molecules that mimic those of host cells, inhibiting
immune recognition and eliciting autoimmunity.
American trypanosomiasis The destruction of host cells is an autoimmune
Infection by Trypanosoma cruzi, the agent that causes phenomenon but whether it is initiated by antigens
American trypanosomiasis or Chagas disease, is often of host or parasite origin or both is not at all clear.
lethal in children and infants. However, in adults, the Other strategies used by T. cruzi to extend its
initial infection often turns into a chronic disease, survival in vertebrate blood include the production of
sometimes after a long interval, causing an illness surface molecules with anti-complement properties,
characterized by megacardia with megacolon, such as T-DAF, gp58/68 and gp160 [31], and the
mega-esophagus and degeneration of the central elimination of immune membrane complexes by
and peripheral nervous systems. A range of surface molecule turnover through the endocytic
immune-system-evasion mechanisms operate pathway [32] and phospholipases that degrade the
during the infection and contribute to both the anchoring glycoproteins [33].
chronicity of the infection and the pathological
changes associated with it. Amoebiasis
The disease is generally associated with Intestinal amoebiasis caused by Entamoeba histolytica
suppression of the IR that becomes more evident as is a worldwide disease. The most relevant characteristic
the parasitemia advances in blood and tissues. of this parasite is its extensive cytolytic capacity.
Surprisingly, phagocytic activity is increased in Amoebic granules contain a battery of aggressive
immunosuppressed animals [23], suggesting that the components, such as hydrolytic enzymes and potent
immunosuppression is due to this increase and not to cysteine proteases, whose secretion contributes to the
the blocking of the mononuclear phagocytic system, damage of host cells and tissues. The proteases can
as has been observed in leishmaniasis and in African also interfere with the humoral immune response by
trypanosomiasis. A similar phenomenon of increased degrading IgA and IgG antibodies [34]. This is
mononuclear phagocytic activity associated with important if we consider that E. histolytica must
important immunosuppression was also noted in overcome the secretory IgA response during
experimental infections with Plasmodium vinckei intestinal colonization and the serum IgG antibodies
[24]. Moreover, T. cruzi evades destruction by when it goes beyond the intestine. Even though
macrophages by escaping from the phagolysosome amoeba trophozoites activate the alternative
and invading non-phagocytic cells as well as by complement pathway [35], they escape the lytic
modulating the pattern of secreted cytokines at effect of complement and the inflammatory
the transcriptional level. In this sense, it has been response when they invade the host’s tissues by
shown that a T. cruzi membrane GPI-anchored acquiring complement-regulating molecules and
mucin (AgC10) [25] can bind to the macrophage inactivating C3a and C5a mediators [36]. In addition,
surface and induce the secretion of IL-1β but not of E. histolytica has a sophisticated strategy to evade
IL-12 or TNF-α, which are considered to be essential antibodies and the antibody-dependent lytic effect
in a protective response against Chagas disease. mediated by complement through a mechanism that
The parasite also promotes the production of allows it to polarize the antibodies deposited on its
IL-10 and transforming growth factor (TGF) β in surface towards the uroid region, where they are
infected macrophages, which inhibit the induction spontaneously eliminated as molecular aggregates
and effects of IL-12 (Ref. [26]). (capping) [37].

http://parasites.trends.com
276 Review TRENDS in Parasitology Vol.18 No.6 June 2002

Tissue invasion by E. histolytica has been host cell to be infected. Two surface molecules of
associated with suppression of cell-mediated Leishmania have been implicated in the attachment
immunity, suggested to be responsible for the acquired and uptake of promastigotes by the host cells, in
protective immunity observed in convalescence addition to the inhibition of the macrophage
cases of extraintestinal amoebiasis, such as amoebic proteolytic processes: the gp63 surface protease and a
liver abscess (ALA). Entamoeba histolytica exerts lipophosphoglycan (LPG). Interestingly, the roles of
different modulatory effects on macrophages and these molecules in immune evasion appear to
T cells, especially on the T helper cell type-1 (Th1) complement each other. During the initial stages of
response. Cellular anergy accompanies the invasion macrophage infection with Leishmania donovani,
of the human liver by E. histolytica [38] and the LPG promotes intracellular survival of promastigotes
intestinal infection of mice [39]. The effect appears to by inhibiting the fusion of the parasite-containing
be produced by a soluble suppressor present in the phagosome with the lysosomes [46]. If the
sera of the infected host that inhibits the production phagolysosome does form, gp63 assumes a protective
of IL-2 even in cells from uninfected animals. It can function by inhibiting degradative phagolysosomal
be reversed by adding exogenous IL-2 or phorbol enzymes [47]. Once the parasites become amastigotes
myristate acetate and ionomycin, indicating a defect inside the macrophage, they adapt to the life inside
at the level of signal transduction [40]. the acid medium of the phagolysosome because the
The cytotoxic capacity of macrophages and their amastigotes are metabolically more active in an acid
potential as antigen-presenting and cytokine- than a neutral environment. Unlike amastigotes,
secreting cells are reduced during the acute phase of which have large amounts of catalase and superoxide
ALA. Entamoeba histolytica trophozoites inhibit the dismutase, the survival of promastigotes (which have
respiratory burst in human macrophages through a less of these enzymes) depends on their capacity to
monocyte locomotion inhibitory factor [41]. In avoid the respiratory burst. Here again, LPG and
addition, the parasite exerts suppression that appears gp63 contribute by blocking the oxidative burst
to be a local event mediated by direct exposure of through abnormal activation of protein kinase C and
macrophages to E. histolytica and its products. Thus, reduction of its translocation to the membrane [48].
the treatment of murine macrophages with amoebic Moreover, LPG is also involved in the direct
antigens in vitro reduces the production of molecules scavenging of oxygen intermediates thanks to its
associated to the I region of the MHC (Ia molecules) structure, which contains repetitive oxidizable
induced by IFN-γ [42]. Because the effect was reversed phosphorylated disaccharide units.
with cyclooxygenase inhibitor, it was suggested that it The parasite also promotes its survival inside
is due in part to prostaglandin E2 (PGE2), probably the macrophage by preventing apoptosis,
produced by E. histolytica or by the macrophage under antigen presentation by MHC molecules and
amoebic induction. PGE2 elevates cAMP levels in responsiveness to cytokines. The stimulation of
macrophages, triggering the phosphokinase A granulocyte–macrophage colony-stimulating factor
pathway, which, in turn, inhibits the expression of Ia and TNF-α has been suggested as possibly
molecules on the macrophage surface and the release responsible for the inhibition of apoptosis [49]. As for
by T cells of Th1 cytokines such as IL-2 and IFN-γ but antigen presentation, in macrophages stimulated by
not of Th2 cytokines [43]. Similarly, macrophages IFN-γ, Leishmania components including
derived from ALA patients constitutively release low glycosylinositolphospholipids inhibit the expression
levels of TNF-α, whereas peritoneal and spleen of the MHC class-I and -II molecules, and peptide
macrophages, and Kupffer cells from infected animals loading to the few MHC molecules produced [50].
do not [44]. Similarly, gp63 from Leishmania major and
Interestingly, it has also been shown that spleen L. donovani cleave CD4 molecules on T cells,
cells from mice inoculated with a surface protein of interfering with the stabilization of the interaction
220 kDa did not proliferate in vitro, although they between antigen-presenting cells and T helper cells
were induced to secrete Th2 cytokines such as IL-4 [51]. In addition, amastigotes internalize and degrade
and IL-10 (Ref. [45]). It might be said that, during class II MHC molecules [52] and downregulate the
E. histolytica invasion, the parasite controls the IR by expression of macrophage co-stimulatory molecules,
modulating the function and set of cytokines released such as the heat-stable antigen and B7-1 [53]. Similar
by macrophages and T cells with the purpose of to E. histolytica, Leishmania induces the release of
increasing its survival within the liver granuloma. PGE2 [54] and TGF β [55], blocking macrophage
functionality. Through LPG, they also control the
Leishmaniasis response of infected macrophages by downregulating
Human leishmaniasis occurs in cutaneous, the expression of the TNF-α receptor [56] and
mucocutaneous and visceral forms, although these inhibiting the chemotaxis of neutrophils and
are not absolute categories. Leishmania lives monocytes [57].
exclusively in mononuclear phagocytes but lacks any The mechanisms developed by this parasite to
specialized mechanism to penetrate cells and evade complement could be among the most
therefore depends on the phagocytic potential of the sophisticated of any parasite. Unlike procyclic

http://parasites.trends.com
Review TRENDS in Parasitology Vol.18 No.6 June 2002 277

promastigotes, metacyclic promastigotes can resist the same results (Table 1). Strategies depend on the
the complement by spontaneously shedding the momentary requirements of the parasite (which in
membrane attack complex (C5b–C9), probably owing turn depend on the factors such as life-cycle stage), on
to the elongation of the LPG molecule on their surface the location of the parasite inside the host (blood,
[58]. The gp63 also protects the parasite through the mucosa, inside a cell) and on the immunological
proteolytic conversion of C3b to C3bi on the parasite status of the microenvironment. In general, the most
surface [59]. Finally, Leishmania protein kinases common evasion mechanisms are those that avoid
can phosphorylate some complement components, confronting parasites with cellular immunity
including C3, C5 and C9, blocking both pathways (macrophages and T cells, especially the Th1
of activation [60]. response) and the antibody-dependent lytic effect of
Cell-mediated immunity based on a Th1 response complement. In an exhibition of sophisticated action,
has been reported to protect against infection with protozoan parasites secrete molecules that either
L. major, whereas susceptibility seems to be related modulate the immune response by controlling the set
to a Th2 response. In this context, it has been found of cytokines produced or directly cause anergy of
that the L. major metacyclic promastigotes do not immune cells by blocking receptors or inhibiting
induce IL-12 production but actively suppress the production of cytokines and their receptors.
transcription of its gene [61]. Because IL-12 is a major The importance of cellular immunity in the control of
physiological promoter of IFN-γ production and protozoa is evident from their effect on T cells and on
Leishmania is highly susceptible to killing by IFN-γ- the function of macrophages, the pillars of this
activated macrophages, this ability to suppress IL-12 kind of immunity. However, despite the fact that
production would be expected to provide a clear parasites manage the host response for survival,
survival advantage to the parasite. Many questions they do it in a sophisticated way that allows the
Acknowledgements still need to be answered about the participation of infected host to live and, in many cases, to struggle
We thank Th1 and Th2 subsets in leishmaniasis. However, the with other infections. Indeed, doing this is an
Isabel Pérez Montford for
her assistance in
data available at present and their application to essential rule for a successful parasite. Critically
translating this the treatment of the disease in humans are promising. evaluating the mechanisms for evading the immune
manuscript. Our work was system and how were they acquired in parallel with
supported by a grant from
Conclusion the evolution of the human immune system will
Consejo Nacional de
Ciencia y Tecnología, Most protozoan parasites share various mechanisms provide insights into the fundamental aspects of the
5078-M9406 (L.O.O.). used to evade the immune response with practically host–parasite relationship.
References 10 Riley, E.M. et al. (1988) Cellular and humoral 18 van der Ploeg, L.H.T. (1987) Control of variant
1 Godal, T. (1996) New dimensions for parasitology responses to Plasmodium falciparum antigens in surface antigen switching in trypanosomes. Cell
in the 21st century. In Parasitology for the 21st Gambian children during and after an attack of 51, 159–161
Century (Özcel, M.A. and Alkan, M.Z., eds), acute falciparum malaria. Clin. Exp. Immunol. 19 Reinitz, D.M. and Mansfield, J.M. (1990) T-cell-
pp. 1–13, CAB International 73, 17–22 independent and T-cell-dependent B-cell
2 Ramasamy, R. (1998) Molecular basis for evasion 11 Riley, E.M. et al. (1989) Suppression of in vitro responses to exposed variant surface glycoprotein
of host immunity and pathogenesis in malaria. lymphoproliferative responses in acute malaria epitopes in trypanosome-infected mice. Infect.
Biochem. Biophys. Acta 1406, 10–27 patients can be partially reversed by Immun. 58, 2337–2342
3 Frevert, U. (1999) Heparan sulphate and indomethacin. Parasite Immunol. 11, 509–517 20 Paulnock, D.M. and Coller, S.P. (2001) Analysis of
RNA-binding motifs in the malaria 12 Cheresh, D.A. et al. (1984) Interaction of an acute macrophage activation in African
circumsporozoite protein. Biochem. Soc. Trans. phase reactant, alpha-acid glycoprotein trypanosomiasis. J. Leukocyte Biol. 69, 685–690
27, 482–487 (orosmucoid), with the lymphoid cell surface. 21 Darji, A. et al. (1996) In vitro simulation of
4 Holder, A.A. et al. (1999) Merozoite surface A model for nonspecific immune suppression. immunosuppression caused by Trypanosoma
protein 1, immune evasion, and vaccines against Immunology 51, 541–548 brucei: active involvement of gamma interferon
asexual blood stage malaria. Parassitologia 41, 13 Srour, E. et al. (1988) Impairment of T helper and tumor necrosis factor in the pathway of
409–414 function by a Plasmodium berghei-derived suppression. Infect. Immun. 64, 1937–1943
5 Reeder, J.C. and Brown, G.V. (1996) Antigenic immunosuppressive factor. J. Protozool. 35, 22 El-Sayed, N.M. and Donelson, J.E. (1997) African
variation and immune evasion in Plasmodium 441–446 trypanosomes have differentially expressed
falciparum malaria. Immunol. Cell Biol. 74, 14 Riley, E.M. et al. (1989) Plasmodium falciparum genes encoding homologues of the Leishmania
546–554 schizont sonicates suppress the GP63 surface protease. J. Biol. Chem. 272,
6 Schofield, L. and Hackett, F. (1993) Signal lymphoproliferative response to mitogens and 26742–26748
transduction in host cells by a antigens in malaria-immune adults. Infect. 23 Ortiz-Ortiz, L. et al. (1976) Enhanced
glycosylphosphatidylinositol toxin of malaria Immun. 57, 3181–3188 mononuclear phagocytic activity during
parasites. J. Exp. Med. 177, 145–153 15 Plebanski, M. et al. (1997) Immune evasion in Trypanosoma cruzi infection in mice. Inter. Arch.
7 Dubois, P. et al. (1988) Structure and function of a malaria: altered peptide ligands of the Allergy Appl. Immunol. 50, 232–237
thymic peptide is mimicked by Plasmodium circumsporozoite protein. Parasitology 115, 24 Greenwood, B.M. et al. (1971)
falciparum peptides. Ann. Inst. Pasteur Immunol. S55–S66 Immunosuppression in murine malaria. I.
139, 557–567 16 Aidoo, M. et al. (1995) Identification of conserved General characteristics. Clin. Exp. Immunol. 8,
8 Mattei, D. et al. (1989) A heat shock-like protein antigenic components for a cytotoxic 467–478
from the human malaria parasite Plasmodium T lymphocyte-inducing vaccine against malaria. 25 de Diego, J. et al. (1997) Alteration of macrophage
falciparum induces autoantibodies. Eur. J. Lancet 345, 1003–1007 function by a Trypanosoma membrane mucin.
Immunol. 19, 1823–1828 17 Rice-Fight, A.C. et al. (1981) Sequence homologies J. Immunol. 159, 4983–4989
9 Daniel-Riveiro, C.T. and Zanini, G. (2000) near the C-termini of the variable surface 26 Silva, J.S. et al. (1998) The role of IL-12 in
Autoimmunity and malaria: what are they doing glycoproteins of Trypanosoma brucei. Nature 294, experimental Trypanosoma cruzi infection. Braz.
together? Acta Trop. 76, 205–221 53–57 J. Med. Biol. Res. 31, 111–115

http://parasites.trends.com
278 Review TRENDS in Parasitology Vol.18 No.6 June 2002

27 Kierszenbaum, F. et al. (1989) Decreased human 40 Campbell, D. et al. (1999) Serum from Entamoeba histocompatibility complex gene products.
IL-2 receptor expression due to a protozoan histolytica-infected gerbils selectively suppresses J. Immunol. 138, 1926–1932
pathogen. Immunol. Today 10, 129–131 T cell proliferation by inhibiting interleukin-2 51 Locksley, R.M. et al. (1993) Helper T cells without
28 Tarleton, R.L. (1988) Trypanosoma cruzi-induced production. J. Infect. Dis. 179, 1495–1501 CD4: control of leishmaniasis in CD4-deficient
suppression of IL-2 production. II. Evidence for a 41 Rico, G. et al. (1995) Cyclic nucleotide changes mice. Science 261, 1448–1451
role for suppressor cells. J. Immunol. 140, induced in human leukocytes by a product of 52 de Souza-Leao, S. et al. (1995) Intracellular
2769–2773 axenically grown Entamoeba histolytica that Leishmania amazonensis amastigotes internalize
29 Garcia, I.E. et al. (1997) Role of membrane-bound inhibits human monocyte locomotion. Parasitol. and degrade MHC class II molecules of their host
IgM in Trypanosoma cruzi evasion from immune Res. 81, 158–162 cells. J. Cell Sci. 108, 3219–3231
clearance. J. Parasitol. 83, 230–233 42 Wang, W. and Chadee, K. (1995) Entamoeba 53 Kaye, P.M. et al. (1994) Deficient expression of
30 Ortiz-Ortiz, L. et al. (1980) Polyclonal B histolytica suppresses gamma interferon-induced co-stimulatory molecules on Leishmania-infected
lymphocyte activation during Trypanosoma cruzi macrophage class II major histocompatibility macrophages. Eur. J. Immunol. 24, 2850–2854
infection. J. Immunol. 124, 121–126 complex Ia molecule and I-Ab mRNA expression 54 Reiner, N.E. and Malelmud, C.J. (1985)
31 Fisher, E. et al. (1988) gp58/68, a parasite by a prostaglandin E2-dependent mechanism. Arachidonic acid metabolism by murine
component that contributes to the escape of the Infect. Immun. 63, 1089–1094 peritoneal macrophages infected with
trypomastigote form of T. cruzi from damage by 43 Betz, M. and Fox, B.S. (1991) Prostaglandin E2 Leishmania donovani: in vitro evidence for
the human alternative complement pathway. inhibits production of TH1 lymphokines but not of parasite-induced alterations in cyclooxygenase
Immunology 65, 299–303 TH2 lymphokines. J. Immunol. 146, 108–113 and lipooxygenase pathways. J. Immunol. 134,
32 Teixeira, R.L. and Santana, J.M. (1989) 44 Wang, W. et al. (1992) Modulation of tumor 556–563
Trypanosoma cruzi: endocytosis and degradation necrosis factor production by macrophages in 55 Barral, A. et al. (1993) Transforming growth
of specific antibodies by parasite forms. Am. J. Entamoeba histolytica infection. Infect. Immun. factor beta as a virulence mechanism for
Trop. Med. Hyg. 40, 165–170 60, 3169–3174 Leishmania braziliensis. Proc. Natl. Acad. Sci.
33 Almeida, I.C. et al. (1994) GPI-anchored 45 Talamas-Rohana, P. et al. (1995) T-cell U. S. A. 90, 3442–3446
glycoconjugates from Trypanosoma cruzi suppression and selective in vivo activation of 56 Corradin, S.B. et al. (1991) Phagocytosis enhances
trypomastigotes are recognized by lytic anti-α- TH2 subpopulation by the Entamoeba histolytica murine macrophage activation by interferon-
galactosyl antibodies from patients with chronic 220-kilodalton lectin. Infect. Immun. 63, gamma and tumor necrosis factor-α. Eur. J.
Chagas’ disease. Braz. J. Med. Biol. Res. 27, 3953–3958 Immunol. 21, 2553–2558
443–447 46 Desjardins, M. and Descoteaux, A. (1997) 57 Frankenburg, S. et al. (1990) Effect of glycolipids
34 Que, X. and Reed, S.L. (1997) The role of Inhibition of phagolysosomal biogenesis by the of Leishmania parasites on human monocyte
extracellular cysteine proteinases in pathogenesis Leishmania lipophosphoglycan. J. Exp. Med. 185, activity. Inhibition by lipophosphoglycan.
of Entamoeba histolytica invasion. Parasitol. 2061–2068 J. Immunol. 145, 4284–4289
Today 13, 190–194 47 Sorensen, A.L. et al. (1994) Leishmania major 58 Puentes, S.M. et al. (1990) Serum resistance of
35 Ortiz-Ortiz, L. et al. (1978) Activation of the surface protease gp63 interferes with the function metacyclic stage Leishmania major
alternative pathway of complement by Entamoeba of human monocytes and neutrophils in vitro. promastigotes is due to the release of C5b–9.
histolytica. Clin. Exp. Immunol. 34, 10–18 Acta Pathol. Microbiol. Immunol. Scand. 102, J. Immunol. 145, 4311–4316
36 Gutierrez, L. et al. (1997) A mechanism of acquired 265–271 59 Brittingham, A. et al. (1995) Role of the
resistance to complement-mediated lysis by 48 Giorgione, J.R. et al. (1996) Transbilayer Leishmania surface protease gp63 in complement
Entamoeba histolytica. J. Parasitol. 83, 234–241 inhibition of protein kinase C by the fixation, cell adhesion, and resistance to
37 Calderón, J. and Avila, E.E. (1986) Antibody- lipophosphoglycan from Leishmania donovani. complement-mediated lysis. J. Immunol. 155,
induced caps in Entamoeba histolytica: isolation Proc. Natl. Acad. Sci. U. S. A. 93, 11634–11639 3102–3111
and electrophoretic analysis. J. Infect. Dis. 153, 49 Moore, K.J. and Matlashewski, G. (1994) 60 Hermoso, T. et al. (1991) Leishmanial protein
927–932 Intracellular infection by Leishmania donovani kinases phosphorylate components of the
38 Ortiz-Ortiz, L. et al. (1975) Cell-mediated inhibits macrophage apoptosis. J. Immunol. 152, complement system. EMBO J. 10, 4061–4067
immunity in patients with amebic abscess of the 2930–2937 61 Carrera, L. et al. (1996). Leishmania
liver. Clin. Immunol. Immunopathol. 4, 127–134 50 Reiner, N.E. et al. (1987) Parasite accessory cell promastigotes selectively inhibit interleukin 12
39 Ghosh, P.K. et al. (1995) Intestinal amebiasis: interactions in murine leishmaniasis. II. induction in bone marrow-derived macrophages
cyclic suppression of the immune response. Leishmania donovani suppresses macrophage from susceptible and resistant mice. J. Exp. Med.
Parasitol. Res. 81, 475–480 expression of class I and class II major 183, 515–526

Trends in Parasitology online


Make the most of your personal subscription:
• High quality printouts (from PDF files)
• Links to other articles, other journals, and cited software and databases

To claim your FREE online access to Trends in Parasitology:


(1) Go to http://www.bmn.com/general/subkey and select
Trends in Parasitology from the list
(2) Enter your own BioMedNet login details when prompted
(if you are not yet a member, joining takes minutes and is FREE)
(3) Follow the instructions on the Trends in Parasitology page under ‘Personal
Subscriber Access’
You only need to register once.
For subsequent visits bookmark: http://journals.bmn.com
If you have any questions e-mail: info@current-trends.com

http://parasites.trends.com

You might also like