Você está na página 1de 21

REVIEWS OF INFECTIOUS DISEASES. VOL. 5, NO.5.

SEPTEMBER-OCTOBER 1983
© 1983 by The University of Chicago. All rights reserved. 0162-0886/83/0505-0008$02.00

The Immunobiology of Leishmaniasis


Richard D. Pearson, David A. Wheeler, From the Divisions of Geographic Medicine and of
Lee H. Harrison, and H. David Kay Rheumatology, Department of Internal Medicine, University
of Virginia School of Medicine, Charlottesville, Virginia

Members of the genus Leishmania are important intracellular pathogens that produce
either cutaneous, mucocutaneous, or visceral disease in many areas of the world. In hu-
mans as well as in other mammals, the parasite is inoculated through the skin as a
flagellated, extracellular promastigote by its arthropod vector, the sandfly. Once in its
mammalian host, the promastigote converts to its amastigote stage, which lacks an ex-
teriorized flagellum and is found solely within mononuclear phagocytes during estab-
lished infection. In vitro, human monocyte-derived macrophages and peritoneal mac-
rophages from several species of rodents can ingest both promastigotes and amas-
tigotes, and they can permit intracellular multiplication of amastigotes only. Although
serum factors may play a role in the pathogenesis of the disease and in protection
against reinfection, the resolution of leishmaniasis is dependent primarily on cell-
mediated immune responses. There appears to be a complicated interplay between cell-
mediated helper and suppressor activities. The outcome of infection in each type of
leishmaniasis depends on the complex and intriguing interaction of virulence factors

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


inherent in the parasite and genetically determined host defense mechanisms.

Leishmania are intracellular protozoal parasites parasite and defining critical aspects of its interac-
that are responsible for significant morbidity and tion with mammalian host defense mechanisms.
mortality in many areas throughout the world.
Leishmaniasis is the general name given to infec-
The Parasite
tion caused by any member of the genus Leish-
mania. Clinical manifestations range from visceral Leishmania are dimorphic protozoa, i.e., their life
disease to one of several forms of cutaneous dis- cycle involves existence in two distinct forms. In
ease, depending on the infecting species. So seri- infected mammalian hosts, the parasite is found
ous is the threat of leishmaniasis in endemic areas almost exclusively within cells of reticuloendothe-
that the World Health Organization has selected it lial origin in its amastigote form, which is 2-3 urn
as one of the six major diseases for study in its in length and oval or round in shape and lacks an
Special Programme for Research and Training in exteriorized flagellum (figure 1). In sandfly vec-
Tropical Diseases. As a result of this emphasis and tors (Phlebotomus species and Lutzomyia species)
recent interest in leishmaniasis, important ad- on the other hand, the parasite converts to and is
vances have been made toward understanding the then transmitted as a flagellated, extracellular
promastigote. Promastigotes have pear- or spindle-
shaped bodies, are 10-15 f.Lm in length and 1.5-3.5
Received for publication June 20, 1982, and in revised form f.Lm in width, and have long unitary flagella (figure
February 22, 1983. 2).
This work was supported by grant no. 1 ROI AI 184802-01 Although minor ultrastructural differences have
from the National Institutes of Health and by grant no. RF
been described [1-7], most investigators find it
79062 from The Rockefeller Foundation.
We are indebted to Dr. Sydney S. Breese, Associate Director difficult, if not impossible, to differentiate species
of the Central Electron Microscope Facility at the University of of Leishmania that cause disease in humans, from
Virginia School of Medicine, for assistance in obtaining elec- one another on the basis of morphology in either
tron micrographs. We are especially grateful to Ms. Anne the amastigote or promastigote stages. Determina-
Groschel and Ms. Susan Davis for their help in preparing the
tion of species type was originally based on a vari-
manuscript.
Please address requests for reprints to Dr. Richard D. Pear- ety of factors such as: (1) the parasite's behavior
son, Division of Geographic Medicine, Box 485, University of in humans; (2) epidemiologic differences asso-
Virginia School of Medicine, Charlottesville, Virginia 22908. ciated with its geographic distribution; (3) the in-

907
908 Pearson et al.

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011

Figure 1. (A) Leishmania donovani amastigotes (arrows) are seen in a touch preparation made from an infected
Syrian hamster spleen (Wright-Giemsa stain). Amastigotes are 2-3 J.tm in diameter and have an eccentrically located
nucleus and a dense-staining rod-shaped kinetoplast. The nuclei (N) of host mononuclear phagocytes are also seen.
The bar represents 10 J.tm. (B) Transmission electron micrograph of L. donovani amastigotes isolated from an infected
hamster spleen showing the parasite nucleus (N), flagellum (F), and kinetoplast (K). A dividing parasite is present.
The bar represents 1 J.tm.

volvement of different animal reservoirs; and (4) pending in some instances on the mammalian host
transmission by different species of sandflies. that is infected. For example, species of Leish-
These traditional approaches to speciation have mania that produce cutaneous disease in humans
proved inadequate. Clinical syndromes overlap can cause visceral disease in rodents [8]. Because
and the manifestations of disease can vary, de- of these difficulties, several biochemical methods
Immunobiology of Leishmaniasis 909

Figure 2. Scanning electron micro-


graph of a Leishmania donovani pro-
mastigote (P) being ingested by a hu-
man mononuclear phagocyte (M) in
vitro. Most of the parasite's flagellum
lies within a phagocytic pseudopod
(arrow). The bar represents 5 (.Am.

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


of speciation have been proposed: isoenzyme de- for limiting the spread of some species to the skin
termination [9-13], buoyant density analysis of and mucous membranes.
nuclear and kinetoplast DNA [10, 13, 14], radio-
respirometry [15], and analysis of antigenic differ-
Visceral Leishmaniasis
ences in promastigotes or in their excreted factors
[13, 16, 17]. While none of these procedures has In visceral leishmaniasis (Leishmania donovani
proved completely reliable in and of itself, deter- complex), amastigotes multiply within mononu-
mination of isoenzyme patterns is currently the clear phagocytes throughout the reticuloendotheli-
most widely used method of speciation. The devel- al system, including the spleen, liver, lymph
opment of species-specific monoclonal antibodies nodes, and bone marrow [29]. Intermittent fever,
[18] or characterization by restriction endonuc- anemia, and pronounced hepatosplenomegaly are
lease digestion of kinetoplast DNA may ultimately common. There is usually progressive weight loss,
provide a superior basis for species identification cachexia, and malaise followed by bacterial super-
and is currently an extremely active area of re- infection, a complication that can result in death.
search. Asymptomatic infections may occur with L. dono-
vani, but the data regarding this question certainly
are not complete [26]. In general, once the stage of
Clinical Manifestations
hepatosplenomegaly is reached, host defense
The clinical syndromes caused by species of Leish- mechanisms are not able to prevent further para-
mania will be briefly outlined in order to provide site multiplication.
a basis for discussion of the host-parasite interac- Although patients with visceral leishmaniasis
tion. Comprehensive reviews of the epidemiology fail to mount delayed-type skin hypersensitivity
and clinical features of leishmaniasis can be found reactions to leishmanial antigen (as measured by
elsewhere [19-28]. As alluded to earlier, various the Montenegro test), they do have markedly ele-
species of Leishmania produce either visceral or vated globulin levels and produce Leishmania-
one of several forms of cutaneous disease. The specific antibodies. Globulin levels as high as 5
distinct tissue tropism of the various leishmanial g/ 100 ml are occasionally observed, but much of
species is poorly understood at present. Sensitivity this globulin is not Leishmania specific [23]. After
to temperature, for example, may be responsible successful treatment with a pentavalent organic
910 Pearson et al.

antimonial agent (e.g., antimony sodium stiboglu- isolated or grouped tubercules appear over or
conate) or an alternative drug, delayed hypersensi- around scars of healed cutaneous ulcers [30].
tivity to leishmanial antigen usually develops These new lesions are associated with the persis-
within several months, antibody levels gradually tence of a few parasites in scars or proximallym-
decline, and there is usually resistance to reinfec- phatic channels.
tion [20]. This series of events has led to specula- In diffuse cutaneous leishmaniasis, a relatively
tion that potentially effective cell-mediated host uncommon syndrome, disease starts as a localized
defense mechanisms are suppressed during active lesion, usually a papule, that does not ulcerate.
visceral leishmaniasis. Satellite lesions subsequently develop around the
A condition known as post-kala-azar dermal initial papule, and organisms may metastasize to
leishmaniasis is sometimes seen as a sequel to distant areas of the skin, often to the face and ex-
treated L. donovani infection. In this syndrome, tremities. The disease runs a protracted course,
widely disseminated cutaneous lesions, which con- but visceral dissemination does not occur. Diffuse
tain L. donovani amastigotes in macrophages, cutaneous leishmaniasis has been described in
develop months to years after successful treatment both the New World [31-34] and the Old World
of visceral leishmaniasis. [35]. Antibodies are demonstrable, whereas de-
layed hypersensitivity responses to leishmanial
antigen are absent. It has been hypothesized that

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


Cutaneous Leishmaniasis
the disease simply represents poor host resistance,
The cutaneous forms of leishmaniasis occur in but it is possible that parasite factors playa role
both the Old World (e.g., Leishmania tropica as well. There is evidence suggesting that, as in vis-
complex) and the New World (e.g., Leishmania ceral leishmaniasis, cell-mediated immune re-
mexicana complex and Leishmania braziliensis sponses against the parasite are suppressed during
complex). Once inoculated by the sandfly into the diffuse cutaneous leishmaniasis [34].
skin, it seems apparent that parasites multiply in Mucocutaneous leishmaniasis is found predom-
macrophages. In classic localized cutaneous leish- inantly in South America, where L. brasiliensis
maniasis, a papule forms and eventually ul- complex is usually implicated as the causative
cerates. Lesions usually heal spontaneously within species. Patients initially develop cutaneous le-
several months, but a flat, atrophic scar remains sions that can be followed months to years later by
as evidence of the disease. The Montenegro skin destructive mucosal lesions. Because the most
test usually becomes positive four to six weeks common sites of mucosal involvement are the
after onset of the lesion. Leishmaniasis recidivans cartilaginous structures of the nose, mouth, and
is a variant of cutaneous leishmaniasis in which pharynx, gross deformities of the face can result

Figure 3. Patient with mucocutane-


ous leishmaniasis who has severe in-
volvement of the face and concomi-
tant destruction of the nasal septum.
(Photograph courtesy of Dr. Anasta-
do de Queiroz Sousa, Universidade
Federal do Ceara, Fortaleza, Brazil).
Immunobiology of Leishmaniasis 911

(figure 3). The percentage of patients with cutan- munobiology of the various forms of leish-
eous lesions who subsequently go on to develop maniasis is not possible, important advances have
mucocutaneous disease is unknown. Isolation of been made toward understanding the parasite's
the organism by direct smear or culture of muco- biology and defining critical aspects of their inter-
sal lesions is difficult. The Montenegro skin test is action with mammalian host defense mechanisms.
positive in most cases, a fact indicating that the These advances are discussed in detail in the sec-
host is capable of mounting cell-mediated immune tions that follow. Attention will focus on recent
responses to leishmanial antigen. progress. The older literature is well summaried
In some respects, the range of histopathological elsewhere [19-24, 27] and will be referred to only
features in the cutaneous forms of leishmaniasis is as it relates to recent studies.
analogous to that of leprosy [19, 27, 33, 36]. At When a Leishmania-infected sandfly attempts
one end of the spectrum lies diffuse cutaneous to take a blood meal from its mammalian host,
leishmaniasis, in which there is little evidence of promastigotes are inoculated. The precise se-
an effective immune response. Vacuolated, heavi- quence of events that follows inoculation has not
ly parasitized macrophages are abundant, but been defined. Although Leishmania exist only
there are few lymphocytes. The cutaneous reac- transiently in the promastigote stage, the initial
tion to leishmanial antigen is negative. The histol- interaction of promastigotes with mammalian
ogy of diffuse cutaneous leishmaniasis is similar to host defense mechanisms is critical to the develop-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


that of lepromatous leprosy, in which there is mas- ment of infection. Based on in vitro studies with
sive bacterial infection of macrophages and little human serum and polymorphonuclear leukocytes,
cell-mediated immune response. At the other end it appears that promastigotes must escape poten-
of the spectrum lies mucocutaneous leishmaniasis. tially lethal humoral and cellular host defense
In the mucosal lesions, there is a pronounced mechanisms in order to gain entrance into macro-
mononuclear cell infiltrate, but few if any para- phages. Once Leishmania have become amasti-
sites can be found. This situation is analogous to gotes within macrophages, resolution of infection
that of tuberculoid leprosy, in which there is an depends predominantly on cell-mediated immune
intense mononuclear infiltrate with few bacteria mechanisms [37-39].
[19, 33].
Treatment of typical cutaneous leishmaniasis
Genetic Control of Resistance to Leishmania
with a pentavalent antimonial drug generally re-
sults in progressive lymphocytic infiltration, a Unfortunately, no data are available about the
decrease in parasite density, and reversion to skin 'genetic control of susceptibility of humans to
test positivity [27]. Local necrosis of the epidermis leishmanial infection. On the other hand, it seems
and ulceration may occur. These findings have led clear that resistance to L. donovani and L. tropica
to speculation that antileishmanial therapy re- is genetically determined in mice [40-46]. Some
duces the parasite burden and permits the expres- inbred strains of mice are susceptible to visceral or
sion of cell-mediated immune mechanisms at cutaneous leishmaniasis, while others are resis-
levels that may be sufficient to resolve the disease. tant.
Resistance to L. donovani. Bradley and co-
workers have reported that resistance to L. don-
Immune Response to Leishmania
ovani is controlled by a single autosomal gene on
Development of clinical leishmaniasis is depen- chromosome 1. The gene has been designated Lsh
dent on both host and parasite factors. The chal- [41,42]. This genetic locus, which controls natural
lenge is to separate invasiveness, pathogenicity, resistance to L. donovani in mice, is closely linked
and parasite survival into intrinsic host and para- to, and probably identical with, the locus that con-
site components [20]- in other words, to deter- trols resistance to Salmonella typhimurium [45].
mine the degree to which the clinical form of leish- Among susceptible strains of mice, some exhibit
maniasis is an expression of the parasite's genetic prolonged infection while in others there is a pro-
constitution (virulence) or the host's innate suscep- gressive decrease in parasite burden late in infec-
tibility and capacity to respond (immunity). Al- tion. This late response is controlled by a genetic
though a comprehensive explanation of the im- locus within or close to the major histocompatibil-
912 Pearson et af.

ity complex (H-2) [43] as well as by genes asso- Early Events in Leishmaniasis: The Interaction of
ciated with minor histocompatibility loci (H-11) Leishmania Promastigotes with Mammalian Host
and with the Ir-2 locus [44]. Even in susceptible Defense Mechanisms
C57Bl mice, Semprevivo et al. [46] have demon- Macrophage-promastigote interactions. Although
strated a spectrum of visceral disease among con- Leishmania exist only transiently as promastigotes
geneic strains. Unfortunately, the mechanisms(s) in their mammalian hosts, the initial interaction of
by which resistance to visceral leishmaniasis is the promastigote stage with cellular and humoral
mediated in the various mouse strains has not yet host defense mechanisms is critical to the develop-
been defined and may differ from strain to strain. ment of infection. In genetically susceptible mam-
Resistance to L. tropica. The capacity of cer- mals, the macrophage appears to serve as a
tain murine strains to be infected with L. tropica sanctuary in which Leishmania promastigotes
is likewise under genetic control [40, 47-53]. escape the potentially adverse effects of serum and
BALBI c mice, which are susceptible, have pro- polymorphonuclear leukocytes and convert to the
gressive local disease followed by disseminated in- amastigote stage.
fection, while C57Bl, mice which are considered Several investigators have performed micro-
resistant, can be infected but display localized, scopic studies of the interaction of promastigotes
self-healing ulcers. Relative resistance to L. tropi- with resident, rodent peritoneal macrophages [55-
ca is controlled by a single autosomal gene dif- 59] and tumor cell lines [60] in vitro and have

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


ferent from Lsh and not involved in the H-2 com- shown that promastigotes are rapidly ingested
plex. Howard et al. [49] have shown - with radia- after macrophages bind them by either their
tion chimeras, which are compatible at the H-2 flagellar or aflagellar poles. Ingestion is followed
locus - that susceptibility to L. tropica is deter- by phagosome-lysosome fusion. Promastigote in-
mined by the descendants of donor hematopoietic gestion can be inhibited by cytochalasin Band cy-
(bone marrow) cells and not by environmental tochalasin D [58, 59], which interfere with poly-
factors. The susceptibility of BALB/c mice to merization of microfilaments in macrophages and
L. tropica appears not to be due to a failure to elicit thereby block the phagocytic mechanism. There is
a delayed hypersensitivity response, but rather to no evidence that promastigotes by themselves can
be the result of induction of a suppressor T cell actively penetrate the macrophage membrane.
population(s) [50]. The findings of Handman There appear to be "recognition" or "binding"
et al. [52] suggest that the susceptibility of strains sites that facilitate the attachment of promasti-
of mice to L. tropica may depend both on a per- gotes to macrophages. Evidence presented by
missive macrophage and on defective T cell recog- Chang [61] indicates that the attachment of L. don-
nition of parasite antigens on the surface of in- ovan; promastigotes to rnacrophages occurs
fected macrophages (for further discussion see via a receptor-ligand interaction. Experiments in
Suppression of Immune Mechanisms below). which promastigotes were treated with glycosi-
Although nothing is known about the genetic dases and competition experiments in which
control of resistance to leishmanial infection in various monosaccharides were used suggest that
humans, Walton and Valverde [54] have observed some of the binding ligands are glycoconjugates.
racial differences in the course and outcome of Chang found that promastigote-macrophage
mucocutaneous leishmaniasis in residents of the binding was sharply reduced by treating either
eastern Andes of Bolivia. They found that extreme parasites or macrophages with trypsin or by incu-
facial mutilation due to mucocutaneous leish- bation with the cation chelator EGTA. Zenian
maniasis is almost exclusively confined to persons [62] has reported that attachment of L. tropica
of African ancestry, even though many more cases promastigotes to murine macrophages is depen-
of leishmaniasis are seen among indigenous dent on various carbohydrates and is inhibited by
Amerinds in the area. Skin testing with leish- EDTA. These studies suggest the existence of a
manial antigen gave greater reactions among glycoprotein receptor(s) or binding site(s) on the
blacks and suggested that mutilation might be due parasite andlor macrophage, the activity of which
to necrosis resulting from an exaggerated immune is dependent on the availability of divalent
response. cations.
Immunobiology of Leishmaniasis 913

A number of investigators have observed that 50


PM 1011
promastigotes can be killed intracellularly after
ingestion in vitro by rodent peritoneal macro- 40
phages [58, 63-65]. This led to speculation that, in
nature, promastigotes might convert to the more ..,
I
resistant amastigote stage within skin fibroblasts Q 30
and that amastigotes released from fibroblasts x
~
might subsequently infect macrophages. Support e, 20
o
for this hypothetical concept is based on the work
of Chang [66], who demonstrated that a species of
10
Leishmania that produced cutaneous infection in
humans (but not L. donovanii could convert from
the promastigote to an amastigote-like state in
15 30 45 60 75 90 lOS 120 I3S 150 165 180
human skin fibroblasts in vitro. However, Leish-
TIME (MINUTES)
mania have not been identified within fibroblasts
in infected animals or humans, and thus a critical Figure 5. Oxidative response of human monocyte-
piece of data necessary to substantiate this theory derived macrophages exposed to Leishmania donovani.
A luminol-enhanced (1.25 x 1O-6M) chemiluminescence
has not been observed. A much more likely hy-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


assay was used to assess the oxidative burst at a promasti-
pothesis is that rodent peritoneal macrophages gote (PM):phagocyte ratio of 10:1 (. .); amasti-
studied in vitro differ from resident macrophages gote (AM):phagocyte ratio of 10:1 (~); or amas-
in the dermis and that conversion of promasti- tigote.phagocyte ratio of 50:1 (0-----0) in the presence
gotes to amastigotes occurs within macrophages in of 10070 nonimmune, heat-inactivated (at 56 C for 30 min)
serum. Mononuclear phagocytes were studied after five
the skin.
days of cultivation in vitro. Each point represents the cor-
rected mean cpm ± SEM per 1 x 105 macrophages,
based on duplicate determinations. Amastigotes were used
after incubation at 37 C for 4 hr to ensure shedding of
residual hamster membranes as documented by electron
l/l 2000
Q) microscopy. Reprinted with permission from Journal of
>.
o Immunology [70].
o
0'
o
ct 1000
... 800 Pulvertaft and Hoyle [55] were the first to ob-
g 'serve that human mononuclear phagocytes ob-
U
::I
C
oc tained from serous exudates in pathological con-
o ditions or from bone marrow aspirates could
~
ingest L. donovani promastigotes; however, these
o
Q investigators neither quantified their observations
nor assessed the fate of intracellular parasites.
Pearson et al. [67] subsequently demonstrated that
human monocyte-derived macrophages cultivated
for five or more days in vitro bind and ingest
48 72 promastigotes (figure 2), permit their conversion
Hours to an amastigote-like state, and support subse-
Figure 4. Leishmanial infection of human monocyte- quent intracellular parasite multiplication (figure
derived macrophages. Peripheral blood mononuclear 4). Their findings have subsequently been con-
phagocytes were cultivated in vitro for five days and then firmed by Murray and Cartelli [68].
infected with Leishmania donovani promastigotes at a Several investigators have assessed the phago-
parasite.macrophage ratio of 20:1 for 2 hr. Each point cytic oxidative response that accompanies inges-
represents the mean ± SEM of cell-associated parasites
per 100total macrophages, based on four or more experi- tion of promastigotes by macrophages. These
ments done in duplicate. Reprinted with permission from studies followed the observation of Wilson et al.
Journal of Immunology [70]. [69] that Toxoplasma gondii failed to trigger a
914 Pearson et al.

phagocytic oxidative burst when ingested by hu- clear phagocytes correlates with the magnitude of
man mononuclear phagocytes, This is not the case the phagocytic oxidative burst and suggest that the
with leishmanial promastigotes, however. Pearson oxidative microbicidal capacity of a mononuclear
et al. [70, 71] and Murray and Cartelli [68] have phagocyte may be an important determinant of its
demonstrated that human mononuclear phago- susceptibility to leishmanial infection.
cytes can undergo an oxidative burst when ex- Conversely, the survival of promastigotes with-
posed to promastigotes (figure 5). Similarly, in human monocyte-derived macrophages, which
Meshnick and Eaton [72] and Murray [73] re- undergo a phagocytic oxidative burst even though
ported that an oxidative burst accompanies the their response is less potent than that of mono-
ingestion of promastigotes by peritoneal macro- cytes or lymphokine-activated macrophages, sug-
phages from several strains of mice, whether or gests that promastigotes possess some means of
not they are genetically susceptible or resistant to self-defense against macrophage microbicidal
Leishmania. mechanisms. The Leishmania have an iron-con-
The survival of leishmanial promastigotes in taining superoxidase dismutase that differs from
human monocyte-derived macrophages probably superoxide dismutases found in mammalian cells
relates in part to the macrophage's relatively low [72]. The leishmanial superoxide dismutase may
intrinsic oxidative capacity, lack of myeloperoxi- represent an adaption for survival in the presence
dase, or both. Peripheral blood monocytes have a of activated oxygen products of macrophages in

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


progressive decrease in oxidative capacity after vivo. However, the unique properties of the leish-
several days of in vitro cultivation [74] and lose manial superoxide dismutase may allow for the
lysosomal myeloperoxidase after four days of cul- development of a specific inhibitor that could be
tivation [75]. It has been postulated that these fac- used for chemotherapy [72].
tors may contribute to the survival of T. gondii Although well-endowed with superoxide dis-
and other microbes [76] within mononuclear mutase [72, 73], promastigotes are relatively de-
phagocytes, and such factors are probably im- ficient in catalase and glutathione peroxidase and
portant in the intracellular survival of Leishmania are sensitive to H 20rperoxidase-halide microbici-
as well. dal mechanisms in vitro [72, 73, 78-81]. Murray
Support for this hypothesis comes from the fol- [73] showed that promastigotes are killed in condi-
lowing observations. Promastigotes survive poor- tions in which 10 nM H 202/min is generated from
ly in monocytes obtained from human blood [68, glucose by glucose oxidase in a phagocyte-free sys-
71]. This is in contrast to the survival of pro- tem. The addition of a peroxidase with an appro-
mastigotes in human monocyte-derived macro- priate halide enhanced killing. Pearson and Steig-
phages that are cultivated for five or more days in bigel [78] also demonstrated killing of promasti-
vitro before exposure to promastigotes and that gotes in the glucose-glucose oxidase system and
have a diminished oxidative microbicidal capacity. found that >50070 of promastigotes were killed
In addition, Murray and Cartelli [68] demon- after 2 hr of exposure to 10-5 M H 20 2 •
strated that human monocyte-derived macro- Furthermore, Gottlieb and Dwyer [82, 83] dem-
phages, which were activated by mitogen- or anti- onstrated that L. donovani promastigotes are
gen (non-leishmanial)-stimulated lymphokines, unique in having acid phosphatase activity distri-
produced more H 202 during ingestion of promas- buted over their entire external surface membrane
tigotes than did control macrophages and, in con- and within their flagellar reservoir region. The
trast to control macrophages, killed ingested pro- physiologic role of surface membrane-bound acid
mastigotes. Finally, Murray [77] showed that phosphatase has not been established. Its activity
promastigotes can survive in a murine macro- may enable the organism to obtain necessary
phage-like tumor cell line that is not capable of nutrients from organic phosphates in the environ-
producing a phagocytic oxidative response. Lym- ment, which would not otherwise be available,
phokine activation of these cells prior to incuba- and thereby contribute to the survival of parasites
tion with promastigotes led to restoration of ox- within the phagolysosomal system of macro-
idative microbicidal mechanisms and resulted in phages. Surface-bound acid phosphatase might
killing of ingested promastigotes. These data indi- even in some way protect against macrophage
cate that the killing of promastigotes by mononu- microbicidal mechanisms.
Immunobiology of Leishmaniasis 915

Interaction of promastigotes with PMNs.


Pearson and Steigbigel [78] have shown that ,
INACTIVATED SERUM

PMNs from healthy human donors can ingest and ~


kill L. donovani promastigotes in vitro. Although ts
some promastigotes were ingested in the absence ~
~
of complement, the presence of complement signi-
ficantly facilitated their uptake. Oxidative micro- ~
bicidal mechanisms appeared to be responsible for
@ 105
~
promastigote killing since (1) the ingestion of l.l.J
promastigotes was associated with a phagocytic ~

oxidative burst; (2) promastigotes were shown to t3


~
be susceptible to oxidative microbicidal mecha-
nisms; and (3) there was no evidence of parasite
4
death in PMNs from a donor with chronic granu- 10
lomatous disease of childhood, a condition in
which leukocytes are unable to produce toxic oxi- 0 30 60
dative metabolites. Further studies are needed to TIME (minutes)
determine the role that human PMNs play in de-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


Figure 6. The effect of fresh and heat-inactivated (at
fense against Leishmania and the manner in which 56 C for 30 min) non immune human serum on promasti-
promastigotes avoid ingestion and killing by these gote viability. The number of viable promastigotes, as
cells. assessed by flagellar motility, was determined during incu-
It might be conjectured that in naturally ac- bation at 37 C in fresh serum, 10070 heat-inactivated se-
quired infection promastigotes are inoculated at a rum, or medium alone. Results are expressed as the mean
effect of 10 sera with brackets enclosing ± SEM. Re-
cutaneous site where complement either is not printed with permission from Journal of Immunology
active or is locally inactivated (possibly by factors [92].
in the saliva of the sandfly). Promastigotes would
thus be partially protected from ingestion by
PMNs, and the likelihood that they would safely sera from North American donors with no history
reach the confines of a macrophage would be en- of exposure to Leishmania are also lethal for
hanced. In contrast to ingestion by PMNs, inges- L. donovani promastigotes (figure 6) [92]. Pear-
tion of promastigotes by mononuclear phagocytes son and Steibigel have studies the mechanism
occurs readily in the absence of immune serum or responsible for the lethal effect of serum obtained
complement. from these nonimmune donors and have found
Lethal effects of serum on promastigotes. that promastigotes in vitro activate the comple-
Humoral factors also serve as a potential defense ment cascade through the classical pathway and
against Leishmania. Several investigators have are killed by the complement membrane attack
found that promastigotes are susceptible to lysis complex (C5B-C9).
by both nonimmune and immune serum from The mode of complement activation, the agglu-
animals [84-86] and humans [87-92]. In 1926 tination of promastigotes exposed to heat-inacti-
Hindle et al. [87], working in China, demon- vated serum (56 C for 30 min), and the loss of
strated that fresh, but not heat-inactivated serum lethal activity when sera were absorbed with
from healthy donors and from patients with vis- promastigotes suggested that antibodies were
ceral leishmaniasis destroyed L. donovani pro- responsible for activating the complement cas-
mastigotes in vitro. Subsequently, Adler [88] and cade. Such antibodies against promastigotes may
Taub [89] in Israel, Lainson and Strangways- have developed in these nonimmune donors as the
Dixon [90] in British Honduras, and Ben Rachid result of exposure to common antigens shared
[91] in Algeria reported that sera from donors who with other protozoa, the mycobacteria [93, 94],
had no history of leishmaniasis killed promasti- other bacteria, or material encountered in the en-
gotes of several Leishmania species. It is possible vironment. For example, infection with one mem-
that these donors had had prior subclinical infec- ber of the family Trypanosomatidae can be ac-
tion and had thus acquired immunity. However, companied by production of cross-reacting anti-
916 Pearson et al.

bodies to other members of the family [19, 95, 96]. Later Events in Leishmaniasis: The Interaction of
This could explain the presence of antileishmanial Leishmania Amastigotes with Mammalian Host
antibodies in donors from areas where trypano- Defense Mechanisms
somes and nonmammalian Leishmania are en-
countered, but it is an unlikely explanation for the At this point we turn our attention to the amasti-
presence of antileishmanial antibodies in North gote stage of Leishmania, which is the stage found
American donors with no known exposure to this in the mammalian host during infection, and to
family of protozoa. the way amastigotes, in contrast to promastigotes,
Another possible source for these cross-reacting interact with host defense mechanisms. Once
antibodies against promastigotes might be natural Leishmania have converted to amastigotes within
antibodies that develop in response to antigens macrophages, cell-mediated immune mechanisms
found on mammalian erythrocytes. There are im- appear to be responsible for controlling subse-
munochemical similarities between leishmanial quent infection, although humoral factors may
antigens and human A-B-O blood group antigens playa role in modulating cell-mediated responses
[97, 98]. There is even some epidemiologic data during infection and possibly even in protecting
suggesting that the distribution of Leishmania the host from reinfection. Studies of cell-mediated
species, as classified by excreted factor serotypes, immune responses during leishmaniasis have in-
may correlate with the frequency of A-B-O blood volved a number of animal models and multiple

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


groups in populations in endemic regions [97, 99]. species of Leishmania. Thus, interpretation of the
The role of antibody, complement, and other findings and generalization to humans is difficult
serum factors in defense against leishmania because the immune response can vary as a func-
promastigotes remains uncertain, however. Serum tion of both the mammalian host and the species
factors probably provide an important but incom- of Leishmania. In general, investigators have
plete barrier against infection. Further efforts are focused on (1) the interaction of amastigotes with
needed to understand the factors that allow para- resident peritoneal macrophages from rodents or
sites to reach safely the confines of the mono- with human phagocytes from peripheral blood
nuclear phagocyte. Some promastigotes may be studied in vitro (2) the role of cell-mediated im-
taken up by macrophages before they can be killed mune mechanisms and macrophage "activation"
by serum, or, alternatively, a subset of promasti- on amastigote survival; (3) the role of immune
gotes may be resistant to the lethal effects of com- suppression during active leishmaniasis; and (4)
plement. Dwyer [100] has shown that promasti- the role of antibody and other humoral factors in
gotes have the ability to cap antibodies; that is, altering the parasite-phagocyte interaction.
antibodies against Leishmania that bind to the Amastigote-phagocyte interactions. Studies of
surface of the parasite can be moved into patches the interaction of host cells with amastigotes have
or a single cap on the parasite's surface and, in focused on tumor cell lines [60, 101-103], poly-
some instances, are subsequently shed. However, morphonuclear and eosinophilic leukocytes [104],
capping is a relatively slow process in relation to resident peritoneal macrophages from laboratory
complement activation, and it is unlikely that animals that are not natural hosts of Leishmania
capping plays a role in protecting promastigotes [105-113], and human peripheral blood mono-
from the lethal effect of serum. Alternatively, nuclear phagocytes after various periods of in
saliva or other factors from the sandfly might coat vitro cultivation [114]. The effects of macrophage
promastigotes and thereby protect them from "activation" on intracellular amastigote survival
antibody and/or complement, or salivary factors will be discussed below.
might even locally inactivate complement. This Canine sarcoma cells have been infected in vitro
possibility has not yet been examined. After the with amastigotes or promastigotes of L. mexicana,
parasite converts to the amastigote stage, it ap- L. tropica, and L. donovani [101, 102], and cer-
pears to be resistant to the lethal effects of nonim- tain murine macrophage/tumor cell lines have
mune serum [84, 92]. also been infected with either L. donovani [103] or
Immunobiology of Leishmaniasis 917

L. mexicana [60]. Once infected, the cell lines can mononuclear phagocytes [114], they multiply in
be used to propagate parasites and to assess the phagolysosomes. In electron microscopic studies
effects of chemotherapeutic agents on intracellu- of rodent peritoneal macrophages infected in
lar amastigotes. vitro, Alexander and Vickerman [106] demon-
Recent reports by Wyler [115] and Klempner et strated fusion of parasitophorous vacuoles con-
al. [116] indicate that attachment of amastigotes taining L. mexicana with secondary lysosomes
to human mononuclear phagocytes is dependent labeled with colloid saccharated iron oxide. Chang
at least in part on amastigote factors. Pretreat- and Dwyer [107, 108] confirmed these studies
ment of amastigotes with cytochalasins, which are using hamster peritoneal macrophages and L. dono-
microfilament inhibitors, decreased the attach- vani amastigotes. They demonstrated fusion of
ment of amastigotes to mononuclear phagocytes secondary lysosomes in parasitophorous vacuoles
in vitro. However, once amastigotes have attached using thorotrast (an electron-dense marker).
to a macrophage, ingestion depends on the macro- Phagosome-lysosome fusion was subsequently
phage's phagocytic apparatus. There is no evi- demonstrated in mice in vivo by Berman et al.
dence that amastigotes have the capacity by them- [113]. In this respect Leishmania resemble Myco-
selves to actively penetrate into macrophages. In bacterium lepraemurium [117], which permits
addition, there is evidence from an animal model phagosome-lysosome fusion in macrophages;
that suggests that there may be subpopulations of however, Leishmania differ from viable Mycobac-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


macrophages that differ in their ability to bind terium tuberculosis [118] and T. gondii [119],
and ingest amastigotes and to support subsequent whose survival within macrophages has been attri-
intracellular parasite growth and replication [111]. buted to their capacity to inhibit the fusion of
Amastigotes are not killed by human monocytes parasitophorous vacuoles with lysosomes. Leish-
[68, 71] or by "nonactivated" monocyte-derived mania also differ from Trypansoma cruzi, which
macrophages [114] in vitro. When ingested by escapes from the phagocytic vacuole and resides in
monocyte-derived macrophages, amastigotes elicit the cytoplasm of the host cell [120].
a phagocytic oxidative response, but it is smaller Leishmania amastigotes must be either resistant
than that elicited by an equal number of promasti- to lysosomal enzymes or somehow protected from
gotes (figure 5). Pearson et al. [70] found that the lysosomal contents within the parasitophorous
response to amastigotes was approximately 10070 vacuole. Greenblatt and co-workers [97, 99, 121-
that to promastigotes as measured in a luminol- 123] have found that amastigotes in vivo and
enhanced chemiluminescence assay. Murray and promastigotes in vitro release a complex poly-
Cartelli [68] reported similar results using an assay saccharide, termed excreted factor, that can en-
for release of H 2 0 2 • The reason(s) why the two hance the growth of amastigotes in nonpermissive
parasite stages elicit different responses is not rodent peritoneal macrophages [121] and of
clear but may relate to differences in motility or promastigotes in culture [122]. It is possible that
size, to stage-specific differences in exposed anti- excreted factor plays a role in protecting intra-
gens, or to differences in attachment of the para- cellular amastigotes from potentially toxic lysoso-
site to the macrophage. Murray [116a] has also mal constitutents.
found that amastigotes contain more catalase and The parasitophorous vacuole that contains
glutathione peroxidase than promastigotes and are Leishmania seems to be part of a phagosome-lyso-
four times more resistant to enzymatically gener- some vacuolar apparatus by which exogenous sub-
ated H 2 0 2 • Once inside macrophages, intracellular stances can be brought into contact with amasti-
Leishmania neither stimulate continuous oxidative gotes via phagocytosis or pinocytosis from the ex-
activity nor do they permanently interfere with tracellular menstruum. When Chang and Dwyer
subsequent oxidative responses to other particu- [108] added thorotrast to macrophages previously
late stimuli [70]. infected with amastigotes, the marker was found
After amastigotes have gained entrance into ro- within macrophage pinosomes, secondary lyso-
dent peritoneal macrophages [106-108] or human somes, the parasitophorous vacuole, and even in
918 Pearson et al.

the parasite's flagellar pocket and cytoplasm. The lymphoid cells when administered concurrently
dynamic phagosome-lysosome vacuolar apparatus [135].
may be the basis for the successful delivery of Further evidence for the role of cell-mediated
pentavalent antimonial compounds to macro- immune responses against Leishmania comes
phages by liposomes [124-126] in experimental from animal models of visceral leishmaniasis.
leishmaniasis. Studies by Skov and Twohy [136, 137], in which
Finally, amastigotes, like promastigotes, are immunosuppressive measures were used in C57Bl/
susceptible to ingestion and killing by human 6J mice, suggested that T cells play the predomi-
PMNs and eosinophils [104]. The role of these nant role in acquired immunity to infection with
phagocytes in preventing amastigotes from gain- L. donovani. Rezai et al. [38] later confirmed in
ing access to uninfected mononuclear phagocytes cell transfer experiments that immunity to L. don-
is unknown. In contrast to promastigotes, amasti- ovani infection was mediated by thymus-depen-
gotes have not been shown to be susceptible to dent lymphocytes. Transfer of immune serum
killing by serum factors [84, 92]. alone did not confer immunity to recipient ani-
Cell-mediated immunity and macrophage "acti- mals. The results from these in vivo animal models
vation." Several animal models and in vitro sys- of visceral leishmaniasis were similar to those
tems have been used to investigate the role of cell- from animal models of cutaneous leishmaniasis in
mediated immunity in host defense against Leish- demonstrating the primary role of cell-mediated

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


mania. Many of these studies have been reviewed immune mechanisms in controlling leishmanial in-
in detail elsewhere [19-23, 27, 127]. The impor- fection.
tance of cell-mediated immune mechanisms was The fate of Leishmania within mononuclear
first suggested by studies involving a nonhuman phagocytes appears to depend on the state of mac-
pathogen, Leishmania enriettii, which produce rophage activation. Miller and Twohy [138] ob-
spontaneously healing cutaneous ulcers in guinea served that L. donovani amastigotes would multi-
pigs [128-133]. Healing of the ulcers was found to ply in vitro in peritoneal macrophages from nor-
be associated with the development of delayed mal mice but not from mice previously infected
hypersensitivity skin reactions as well as in vitro with the parasite. Their findings suggested that
lymphocyte proliferative responses to leishmanial macrophages became "activated" during infection
antigen. Although specific antibodies against to inhibit multiplication of intracellular parasites.
Leishmania were produced during infection, they Smrkovski and Larson [139] found that mice
did not result in resolution of the ulcers. Suppres- immunized with BCG were protected against a
sion of cell-mediated immune responses by anti- subsequent challenge with L. donovani amasti-
serum against lymphocytes increased the severity gotes. Such protection could be abrogated, how-
of infection [132, 133], an observation suggesting ever, by treatment with cortisone [140]. In addi-
that the lymphocytes played an important role in tion, L. donovan i-infected mice that received
controlling leishmanial infection. BCG had decreased numbers of amastigotes in
Preston et al. [134, 135] subsequently studied their livers and spleens. The data suggested that
the role of cell-mediated immunity in an experi- macrophages were nonspecifically activated by
mental model of cutaneous leishmaniasis in which BCG to kill intracellular L. donovani, but, be-
CBA mice were infected with a human pathogen, cause species of Leishmania and Mycobacterium
L. tropica. They demonstrated, using thymecto- may have antigenic similarities [93, 94], a cross-
mized, irradiated mice, that the healing of cutane- reacting, antigen-specific response could not be
ous ulcers required an intact T cell system [134]. In excluded. These findings with L. donovani are in
addition, they found that lymphoid cells obtained contrast to those of Grimaldi et al. [141], who
from healed CBA mice or from spleen, peritoneal found that neither BCG nor the immunostimulant
exudates, or lymph nodes conferred protection on levamisole resulted in resolution of cutaneous
recipient syngeneic mice challenged with L. tro- lesions or in production of a hyperallergic reaction
pica. Serum from healed mice was not protective in C3H mice with cutaneous leishmaniasis caused
by itself, but it did enhance the protective effect of by L. mexicana.
Immunobiology of Leishmaniasis 919

The available in vitro data also also indicate "activate" macrophage in vitro to kill intracellular
that the intracellular fate of amastigotes depends amastigotes. Thus, in animal models and in vitro
on the capacity of T cells to activate macrophages. studies of cutaneous and visceral leishmaniasis,
MaueI et al. [142] and Buchmiiller and Mauel intracellular killing of amastigotes appears to be
[143] demonstrated that isolated mouse peritoneal dependent on activation of macrophages by lym-
macrophages endocytize L. enriettii amastigotes in phokines.
vitro and permit their intracellular survival. How- The role of cytotoxic mononuclear cells (such as
ever, cocultivation of infected macrophages with "natural killer" cells) in protection against infec-
syngeneic splenic lymphocytes that had been stim- tion with Leishmania is uncertain. In studies by
ulated either by allogeneic cells in mixed lympho- Bray and Bryceson [128] and Bryceson et al. [129],
cyte culture or by the mitogen concanavalin A mononuclear cells appeared to exert a direct cyto-
(Con A) resulted in intracellular parasite killing. toxic effect against guinea pig macrophages that
In addition, exposure to Con A-stimulated lym- contained amastigotes of L. enriettii or that were
phokines produced a concomitant increase in the coated with leishmanial antigen. However, these
oxidative capacity of macro phages after phagocy- findings were not confirmed by others [148].
tosis [79]. Similar results have been reported by In some instances, the susceptibility of mice to
Murray (l16a] and Haidaris and Bonventre [143a] leishmanial infection may be due to inadequate
with L. donovani. These investigators have hy-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


presentation of leishmanial antigens by macro-
pothesized that the destruction of parasites within phages to lymphocytes. Handman et al. [52] dem-
activated macrophages might be mediated by en- onstrated that macrophages from both an L. tropi-
hanced oxidative microbicidal mechanisms. ca-susceptible strain of mice, BALBIc, and a
The model of Mauel et al. [142, 143] has been relatively resistant strain, CBA/H, had leish-
criticized because L. enriettii does not multiply manial antigens on their surface after incubation
within murine macrophages in vitro. This was not with crude antigen preparations, but only BALBI
a problem, however, in the studies of Nacy et al. c macrophages (in contrast to CBA/H macro-
[144], who defined conditions for the replication phages) failed to sensitize syngeneic recipient cells
of L. tropica amastigotes in mouse peritoneal to produce delayed hypersensitivity responses to
macrophages in vitro and demonstrated that the antigen. This failure seemed to be due to re-
murine macrophages that were preexposed to anti- duced functional expression of H-2 antigens on
gen-stimulated lymphokines were less likely to the surface of BALB/c macrophages. As the
become infected, and that lymphokine-exposed authors noted, however, this cannot be the sole
cells that became infected inhibited replication of explanation for the variation in susceptibility of
intracellular amastigotes. Macrophages that were mice to L. tropica, since other L. tropica-resistant
first infected with amastigotes and then treated mouse strains (e.g., NZB and C57BI/6), in con-
with lymphokines exhibited potent microbicidal trast to CBA/H mice, have macrophages that fail
activity against intracellular amastigotes. Similar to support intracellular parasite multiplication.
results were obtained by Handman and Burgess The data raise the possibility that the genetic sus-
[145] using murine macrophages that were treated ceptibility of mice to L. tropica is associated with
with granulocyte-macrophage colony-stimulating both a permissive macrophage and defective T cell
factor and exposed to L. tropica and by Haidaris recognition of parasite antigens on the surface of
and Bonventre [146] who studied the interaction infected macrophages [52].
of L donovani with lymphokine-activated murine The relevance of these findings to infection in
macrophages. In the latter study, amastigotes humans remains conjectural. Berman et al. dem-
were killed within macrophages only when the onstrated that human monocyte-derived macro-
"activated" state was maintained by daily addition phages, which support intracellular amastigote
of lymphokines for several days. Finally, Chang multiplication in vitro [114], have leishmanial
and Chiao [147] showed that L. donovani infec- antigens on their surfaces [149]. Murray and Car-
tion of immunocompetent mice elicits the produc- telli [68] recently showed that human mononu-
tion of soluble splenic lymphocyte factors that can clear phagocytes could be activated in vitro by
920 Pearson et al.

mitogen-stimulated lymphokines to kill L. dono- sis and 11 patients who had been successfully
vani amastigotes. Further studies are needed to treated one or more years before. In contrast to
define the critical interaction between human the treated patients, infected patients were unre-
macrophages (infected with Leishmania) and sponsive to leishmanial antigen. However, three
lymphocytes in order to determine if there is a of four infected patients became responsive to
genetic or parasite-induced defect in antigen pre- leishmanial antigen when retested several weeks
sentation to subpopulations of macrophage-acti- after successful chemotherapy. The lack of a
vating T cells in the progressive forms of leish- proliferative response to leishmanial antigen could
maniasis. not be attributed to reduced numbers of circulat-
Suppression of immune mechanisms. Clinical ing T cells or to the inhibitory effect of monocytes
experience with human visceral leishmaniasis and or serum factors. Neither infected, treated, nor
diffuse cutaneous leishmaniasis suggests that cell- control Brazilian patients had depressed responses
mediated immune mechanisms may be suppressed to PHA, a fact suggesting that immune suppres-
during active disease. In both syndromes, delayed sion during South American visceral leishmaniasis
hypersensitivity responses to leishmanial antigen is relatively antigen specific.
are absent during infection, but in many patients There is also evidence of broader immune de-
they develop following successful antileishmanial pression during human leishmaniasis. Patients
chemotherapy. The best evidence for immune sup- suffering from Indian visceral leishmaniasis have

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


pression during human leishmaniasis comes from been shown to have depressed lymphocyte respon-
the work of Peterson et al. [32], who studied four siveness to PHA [152]. Some patients in Ethiopia
patients from the Dominican Republic with dif- with diffuse cutaneous leishmaniasis demon-
fuse cutaneous leishmaniasis caused by a newly strated impaired sensitization to dinitrochloroben-
identified strain of Leishmania and found that zene, but they simultaneously had normal delayed
adherent cells from peripheral blood could inhibit hypersensitivity responses to lepromin and tuber-
lymphocyte-proliferative responses to leishmanial culin [153]. Serebryalov et al. [22] have reportedly
antigens. None of the patients had delayed cutane- found that patients infected with L. tropica before
ous reactions or lymphocyte-proliferative re- the third inoculation of diphtheria-pertussis-
sponses to leishmanial antigens, but they did have tetanus vaccine have a decreased antibody re-
normal proliferative responses to mitogens and to sponse to diphtheria. The significance of these ob-
other antigens, so there seemed to be antigen- servations is uncertain because of the variations in
specific suppression in these patients. The total tests and procedures used.
numbers of lymphocytes and T cells in these pa- Evidence from animal models further supports
tients were normal. Decreasing the number of the hypothesis that immune responses in mammals
adherent mononuclear cells by passage over nylon are suppressed during diffuse cutaneous and
wool or addition of the prostaglandin inhibitor visceral leishmaniasis. L. donovani-infected ham-
indomethacin permitted expression of lymphocyte sters were reported to have a diminished capability
responses to leishmanial antigens, thereby suggest- to reject skin homografts [154] and to produce
ing the presence of a population of suppressor antibodies against chicken ovalbumin [155]. Perez
macrophages. In contrast to cells from patients et al. [156] studied the course of cutaneous leish-
with untreated diffuse cutaneous leishmaniasis, maniasis in sensitive (BALB/c) and less suscep-
lymphocytes from patients with localized cutan- tible (C57BI/6) mice. They found that infected
eous leishmaniasis undergo blastogenic responses BALBI c mice exhibited enhanced responses to
to leishmanial antigen during the course of infec- Con A throughout the course of infection, even
tion [150]. though they remained unresponsive to leishmanial
The findings are similar in human visceralleish- antigen. They also reported that the proliferative
maniasis, although a suppressor cell population responses to PHA and lipopolysaccharide (LPS)
has not yet been identified. Carvalho et al. [151] were depressed. On the other hand, lymphocytes
studied the proliferative response of peripheral from C57B1/6 mice had increased DNA prolifera-
blood mononuclear cells to leishmanial antigen tive responses to leishmanial antigen and to the T
and phytohemagglutinin (PHA) in 14 Brazilian cell mitogens PHA and Con A. In subsequent
patients with acute, untreated visceral leishmania- studies, Perez et al. [157] demonstrated suppres-
Immunobiology of Leishmaniasis 921

sion of the IgM plaque-forming cell response to 27, 161]. In general, the presence and magnitude
sheep erythrocytes in C57B1/6 mice infected with of antibody responses in humans are inversely
L. mexicana at a time during infection when pro- related to the extent and progression of disease.
tective immunity to a challenge infection and de- As previously discussed, cell-mediated immune
layed hypersensitivity responses to parasite anti- mechanisms appear to be more effective in con-
gens were apparently unaffected. During the pe- trolling and limiting the course of infection.
riod of maximal immunodepression, spleen cells The situation is similar in animal models of
from infected mice inhibited the plaque-forming leishmaniasis, in which transfer of immune serum
cell response of spleen cells from uninfected mice. has not protected nonimmune recipients from in-
Scott and Farrell [158] observed depressed proli- fection [38, 39]. In addition, Howard et al. [159]
ferative responses to Con A, PHA, and LPS in found that irradiation of L. tropica-infected mice,
BALB/c mice infected with L. tropica. They iden- at levels that ablated antibody production but not
tified an adherent cell population that had the T-suppressor lymphocytes, had no effect on the
capacity to suppress the in vitro response of course of infection. Data from Biozzi high- and
normal spleen cells to Con A. Indomethacin low-responder mice likewise suggested that the
abrogated the suppression. Although the cell pop- presence of antibody against Leishmania did not
ulation responsible for suppression was not result in resolution of infection. Biozzi et al. [162]
specifically identified, the data suggested that used selectivebreeding techniques to develop high-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


macrophages were responsible. and low-responder lines of mice to natural mul-
In contrast, the findings of Howard et al. [48, tideterminant immunogens (e.g., sheep and pi-
50] suggest that the susceptibility of BALBI c mice geon erythrocytes or flagellar and somatic anti-
to infection with L. tropica is due to induction of gens of salmonellae, bovine serum albumin, and
suppressor T lymphocytes that impair a potential- rabbit gamma globulin). Antibody levels are high
ly effective, delayed hypersensitivity response. In and macrophage activity low in the Biozzi high
their model, delayed skin hypersensitivity was de- responders. The converse is true in low respond-
tectable early in infection, but later it was sup- ers. During L. tropica infection, high responders
pressed. When infected mice were subjected to develop large, progressive ulcerating lesions and
sublethal irradiation at levels known to deplete the have high titers of specific antibody. In low re-
precursors of suppressor T lymphocytes, infection sponders the lesions are small and heal sponta-
resolved at the same rate as in genetically resistant neously, while antibody titers remain low [163].
mice, instead of progressing to the expected fatal The mechanism(s) responsible for the spontaneous
outcome [159]. When irradiated mice were recon- resolution of cutaneous lesions during the immune
stituted with normal BALB/c T cells, inexorable phase in this model appears to be independent of
progression of the disease again occurred. Similar- the levels of humoral antibody. As is the situation
ly, in studies done by Mitchell et al. [160] in which in humans, the antibody response correlates in-
hypothymic BALBI c nude mice were reconsti- versely with the degree and progression of disease.
tuted with spleen cell suspensions from normal Even though the presence of antibody against
BALB/c mice, T cell-dependent immune re- Leishmania does not result in resolution of viscer-
sponses appeared to contribute to the chronicity al, diffuse cutaneous, or mucocutaneous leish-
of infection and to inhibit healing of lesions. maniasis in humans or in experimental animals,
On the basis of the evidence from mouse models serum factors can influence the interaction of
and from humans it appears that the outcome of Leishmania with host macrophages and may mod-
leishmaniasis depends on the complex interplay of ulate cell-mediated immune responses during in-
helper and suppressor mononuclear cell popula- fection, perhaps by blocking or "masking" impor-
tions, but the exact nature of the suppressor cell tant antigens. Chang [164] and Herman [165] have
populations remains to be defined. assessed the effects of serum on the interaction of
The role of humoral factors. Markedly ele- Leishmania with phagocytes in vitro. Chang found
vated immunoglobulin levels and antibodies to that rabbit antibodies against amastigotes actually
Leishmania are present in human visceral leish- inhibited the attachment of amastigotes to human
maniasis, and antibodies are present in cases of PMNs and monocytes. Herman studied the effects
cutaneous leishmaniasis in its various forms [19- of serum from C57B1/6J mice that were rein-
922 Pearson et al.

fected with L. donovani 60 days after an initial in- currently receive mononuclear cells from donors
oculation of the parasite. He found that serum infected with Leishmania. It is possible that circu-
obtained 10 to 11 days after reinfection contained lating immune complexes, lymphokines, or other
cytophilic antibodies and opsonins for L. dono- serum constituents in addition to antibodies
vani amastigotes and promastigotes that resulted against Leishmania are responsible for this effect.
in enhanced binding and ingestion of parasites by Desjeux et al. [166] identified circulating im-
elicited and "activated" mouse peritoneal macro- mune complexes and antibodies against IgG, pos-
phages. Theoretically, enhanced attachment of sibly rheumatoid factors, in patients with severe
amastigotes to macrophages could be either detri- mucocutaneous leishmaniasis. There was a close
mental or beneficial to the host, depending on the correlation between the titers of circulating im-
intracellular fate of the parasite. The fact that cy- mune complexes and of antibodies against IgG.
tophilic and opsonic activity were evident at a time Antibodies against IgG were also identified by
when spleen and liver parasite burdens were de- Houba and Allison [167] in the sera of African pa-
creasing (between 10 and 24 days of infection) sug- tients with visceral leishmaniasis. More recently,
gested that humoral factors facilitated parasite up- circulating immune complexes and antibodies
take and subsequent destruction. In contrast, in against IgG (rheumatoid factors) have been identi-
the presence of serum obtained from mice 24 days fied in Brazilian patients [168, 169] with South
after reinfection, the percentage of macrophages American visceral leishmaniasis. In a study of 29

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


able to ingest parasites was decreased to control patients hospitalized with visceral leishmaniasis in
levels or significantly below. northeastern Brazil, Pearson et al. [169] found
Herman's and Chang's data indicate that hu- that 68070 had circulating immune complexes by a
moral factors can alter the parasite-phagocyte in- C1q binding assay and 93070 had rheumatoid fac-
teraction and that the presence and functional tors. The role that rheumatoid factors, circulating
capacity of antibodies against Leishmania may immune complexes, and other serum factors play
vary during the course of infection. Their findings in modulating immune responses during human
also demonstrate that serum from animals in- leishmaniasis deserves further investigation.
fected with Leishmania can actually inhibit the at-
tachment of parasites to phagocytes. As pre-
viously noted, Leishmania are able to attach to References
macrophages in the absence of antibody and com- 1. Sanyal AB, Sen Gupta Pc. Fine structure of Leishmania
plement. Protein or glycoprotein molecules are in dermal leishmanoid. Trans R Soc Trop Med Hyg
thought to be important in this interaction. Anti- 1967;61:211-6
2. Aleman C. Finestructure of cultured Leishmania brasil-
body against Leishmania may inhibit the attach-
iensis. Exp Parastiol 1969;24:259-64
ment of Leishmania to macrophages by masking 3. Pham TD, Azar HA, Moscovic EA, Kurban AK. The ul-
important surface molecules and may thereby trastructure of Leishmania tropica in the oriental sore.
shift the mechanism of parasite attachment from Ann Trop Med Parasitol 1970;64:1-4
a non-antibody-mediated, spontaneous parasite- 4. Gardener Pl, Howells RE, Molyneux DH. Ultrastructure
of Leishmania amastigotes. Trans R Soc Trop Med
phagocyte interaction to one mediated by Fe re-
Hyg 1973;67:23
ceptors. Alternatively, because Leishmania amas- 5. Shaw 11, Lainson R. Leishmaniasis in Brazil. XI. Obser-
tigotes and promastigotes have the capacity to cap vations on the morphology of Leishmania of the brazil-
surface antibody [100], they could theoretically iensis and mexicana complexes. 1 Trop Med Hyg 1976;
defend themselves by capping or shedding anti- 79:9-13
6. Veress B, Abdalla RE, EI Hassan AM. Electron micro-
gen-antibody complexes, which might then block
scope investigations on leishmaniasis in the Sudan. I.
Fe receptors on phagocytes. Morphometric studies on Leishmania amastigotes in
Finally, it seems clear that serum factors may various forms of human leishmaniasis. Ann Trop Med
play an important role in modulating cell-me- Parasitol 1980;74:421-6
diated immune responses during infection. MaueI 7. Abdalla RE. Parasites in Sudanese cutaneous and muco-
sal leishmaniasis. Ann Trop Med Parasitol 1982;76:
and Behin [37], Preston and Dumonde [135], and
299-307
Poulter [39] found that sera from convalescent or 8. Garnham PCC. Th.e genus Leishmania. Bull WHO 1971;
healed animals can augment the level of adoptive 44:477-89
immunity expressed in recipient animals that con- 9. Ebert F. Charakterisierung von Leishmania donovani-
Immunobiology of Leishmaniasis 923

Stammen mit der Disk-Elektrophorese. Z Tropenmed 30. Ramos e Silva J, Paes de Oliviera Netto M. Leishmanids.
Parasitol 1973;24:517-24 Int J Dermatol 1973;12:104-9
10. Gardener PJ, Chance ML, Peters W. Biochemical taxo- 31. Lainson R, Shaw JJ. Leishmaniasis of the New World:
momy of Leishmania. II: Electrophoretic variation of taxonomic problems. Br Med Bull 1972;28:44-8
malate dehydrogenase. Ann Trop Med Parasitol1974; 32. Petersen EA, Neva FA, Oster CN, Diaz HB. Specific in-
68:317-25 hibition of lymphocyte-proliferation responses by ad-
11. Brazil RP. Electrophoretic variation of the enzyme phos- herent suppressor cells in diffuse cutaneous leishmani-
phoglucomutase in different strains of Leishmania. asis. N Engl J Med 1982;306:387-92
Ann Trop Med Parasitol 1978;72:289-91 33. Convit J, Kerdel-Vegas F. Disseminated cutaneous leish-
12. Schnur LF, Chance ML, Ebert F, Thomas SC, Peters W. maniasis. Innoculation to laboratory animals, electron
The biochemical and serological taxonomy of visceral- microscopy and fluorescent antibodies studies. Arch
izing Leishmania. Ann Trop Med Parasitol 1981;75: Dermatol 1965;91:439-47
131-44 34. Convit J, Pinardi ME, Rondon AJ. Diffuse cutaneous
13. Rassam MB, AI-Mudhaffar SA, Chance ML. Isoenzyme leishmaniasis. A disease due to an immunological de-
characterization of Leishmania species from Iraq. Ann fect of the host. Trans R Soc Trop Med Hyg 1972;66:
Trop Med Parasitol 1979;73:527-34 603-10
14. Chance ML, Peters W, Shchory L. Biochemical taxon- 35. Bryceson ADM. Diffuse cutaneous leishmaniasis in Ethi-
omy of Leishmania. I: Observations on DNA. Ann opia. I. The clinical and histological features of the dis-
Trop Med Parasitol 1974;68:307-16 ease. Trans R Soc Trop Med Hyg 1969;63:708-37
15. Decker JE, Schrot JR, Levin GV. Identification of Leish- 36. Manson-Bahr PEC, Winslow DJ. Cutaneous leishmani-
mania ssp. by radiorespirometry. J Protozool 1977; asis. In: Marcial-Rojas RA, ed. Pathology of protozoal

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


24:463-70 and helminthic diseases. Baltimore: Williams and Wil-
16. Schnur LF, Zuckerman A. Leishmanial excreted factor kins 1971:97-114
(EF) serotypes in Sudan, Kenya and Ethiopia. Ann 37. MaueI J, Behin R. Cell-mediated and humoral immunity
Trop Med Parasitol 1977;71:273-94 to protozoan infections with special reference to leish-
17. Schnur LF, Zuckerman A, Greenblatt CL. Leishmanial maniasis. Transplant Rev 1974;19:121-46
serotypes as distinguished by the gel diffusion of fac- 38. Rezai HR, Farrell J, Soulsby EL. Immunological re-
tors excreted in vitro and in vivo. Isr J Med Sci 1972;8: sponses of L. donovani infection in mice and signifi-
932-42 cance of T cell in resistance to experimentalleishmani-
18. Pratt DM, David JR. Monoclonal antibodies that distin- asis. Clin Exp Immunol 1980;40:508-14
guish between New World species of Leishmania. Na- 39. Poulter LW. Mechanisms of immunity to leishmaniasis.
ture 1981;291:581-3 I. Evidence for a changing basis of protection in self-
19. Turk JL, Bryceson ADM. Immunological phenomena in limiting disease. Clin Exp Immunol 1980;39:14-26
leprosy and related diseases. Adv Immunol 1971;13: 40. Cox FEG. Leishmaniasis and mouse genetics. Nature
209-66 1981 ;291:111-12
20. Heyneman D. Immunology of leishmaniasis. Bull WHO 41. Bradley DJ, Taylor BA, Blackwell J, Evans EP, Freeman
1971;44:499-514 J. Regulation of Leishmania populations within the
21. Bray RS. Leishmaniasis in the Old World. Br Med Bull host. III. Mapping of the locus controlling susceptibili-
1972;28:39-43 ty to visceral leishmaniasis in the mouse. Clin Exp Im-
22. Bray RS. Leishmania. Ann Rev Microbiol 1974;28: munol 1979;37:7-14
189-217 42. Bradley DJ. Regulation of Leishmania populations with-
23. Zuckerman A. Parasitological review. Current status of in the host. II. Genetic control of acute susceptibility of
the immunology of blood and tissue protozoa. I. Leish- mice to Leishmania donovani infection. Clin Exp Im-
mania. Exp Parasitol 1975;38:370-400 munol 1977;30:130-40
24. Jones TC. Interactions between murine macrophages and 43. Blackwell J, Freeman J, Bradley D. Influence of H-2
obligate intracellular protozoa. Am J Pathol 1981; complex on acquired resistance to Leishmania dono-
102:127-32 vani infection in mice. Nature 1980;283:72-4
25. Mahmoud AAF, Warren KS. Algorithms in the diagnosis 44. DeTolla LJ Jr, Semprevivo LH, Palczuk NC, Passmore
and management of exotic diseases. XXIV. Leish- HC. Genetic control of acquired resistance to visceral
maniases. J Infect Dis 1977;136:160-3 leishmaniasis in mice. Immunogenetics 1980;10:353-61
26. Marsden PD. Current concepts in parasitology: leish- 45. Plant JE, Blackwell JM, O'Brien AD, Bradley DJ, Glynn
maniasis. N Engl J Med 1979;300:350-2 AA. Are the Lsh and Ity disease resistance genes at one
27. Haghighi P, Rezai HR. Leishmaniasis: a review of se- locus on mouse chromosome 11 Nature 1982;297:510-1
lected topics. Pathol Annu 1977;12(Pt. 2):63-89 46. Semprevivo LH, DeTolla LJ, Passmore HC, Palczuk
28. Chance ML. The six diseases of WHO: leishmaniasis. Br NC. Spectral model of leishmaniasis in congenic strains
Med J 1981;283:1245-7 of mice. J Parasitol 1981;67:8-14
29. Winslow DJ. Kala-azar (visceral leishmaniasis). In: Mar- 47. Kellina 01. 0 razlichiiakh v chuvstvitel'nosti inbrednykh
cial-Rojas RA, ed. Pathology of protozoal and hel- mysheiraznykh liniik Leishmania tropica major. Med
minthic diseases. Baltimore: Williams and Wilkins, Parazitol (Mosk) 1973;42:279-85
1971:86-96 48. Howard JG, Hale C, Chan-Liew WL. Immunological
924 Pearson et al.

regulation of experimental cutaneous leishmaniasis. I. mania enriettii within murine macrophages. Experi-
Immunogenetic aspects of susceptibility to Leishmania entia 1978;34:598-9
tropica in mice. Parasite Immunol 1980;2:303-14 66. Chang K-P. Leishmania infection of human skin fibro-
49. Howard JO, Hale C, Liew FY. Oenetically determined blasts in vitro: absence of phagolysosomal fusion after
susceptibility to Leishmania tropica infection is ex- induced phagocytosis of promastigotes, and their intra-
pressed by haematopoietic donor cells in mouse radia- cellular transformation. Am J Trop Med Hyg 1978;
tion chimaeras. Nature 1980;288:161-2 27:1084-96
50. Howard JO, Hale C, Liew FY. Immunological regulation 67. Pearson RD, Romito R, Symes PH, Harcus JL. Interac-
of experimental cutaneous leishmaniasis. III. Nature tion of Leishmania donovani promastigotes with hu-
and significance of specific suppression of cell-medi- man monocyte-derived macrophages: parasite entry,
ated immunity in mice highly susceptible to Leishmania intracellular survival, and multiplication. Infect
tropica. J Exp Med 1980;152:594-607 Immun 1981;32:1249-53
51. Djoko-Tamnou J, Leclerc C, Modabber F, Chedid L. 68. Murray HW, Cartelli D. Killing of intracellular Leish-
Studies on visceral Leishmania tropica infection in mania donovani by human mononuclear phagocytes:
BALB/c mice. I. Clinical features and cellular changes. evidence for oxygen dependent and indpendent leish-
Clin Exp Immunol 1981;46:493-8 manicidal activity. J Clin Invest 1983, in press
52. Handman E, Ceredig R, Mitchell OF. Murine cutaneous 69. Wilson CB, Tsai V, Remington JS. Failure to trigger the
leishmaniasis: disease patterns in intact and nude mice oxidative metabolic burst by normal macrophages.
of various genotypes and examination of some differ- Possible mechanism for survival of intracellular patho-
ences between normal and infected macrophages. Aust gens. J Exp Med 1980;151:328-46
J Exp BioI Med Sci 1979;57:9-29 70. Pearson RD, Harcus JL, Symes PH, Romito R, Dono-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


53. Behin R, Mauel J, Sordat B. Leishmania tropica: patho- witz OR. Failure of the phagocytic oxidative response
genicity and in vitro macrophage function in strains of to protect human monocyte-derived macrophages from
inbred mice. Exp Parasitol 1979;48:81-91 infection by Leishmania donovani. J Immunol 1982;
54. Walton BC, Valverde L. Racial differences in espundia. 129:1282-6
Ann Trop Med Parasitol 73:23-29, 1979 71. Pearson RD, Harcus JL, Donowitz OR. Survival of
55. Pulvertaft RJV, Hoyle OF. Stages in the life-cycle of Leishmania donovani in human monocytes [abstract
Leishmania donovani. Trans R Soc Trop Med Hyg no. 26]. In: Program and abstracts of the 22nd Inter-
1960;54:191-6 science Conference on Antimicrobial Agents and Che-
56. Miller HC, Twohy OW. Infection of macrophages in cul- motherapy.Washington, D.C.: American Society for
ture by leptomonads of Leishmania donovani. J Proto- Microbiology, 1982.
zool 1967;14:781-9 72. Meshnick SR, Eaton JW. Leishmanial superoxide dismu-
57. Akiyama HJ, Haight RD. Interaction of Leishmania tase: a possible target for chemotherapy. Biochem Bio-
donovani and hamster peritoneal macrophages. A phys Res Commun 1981;102:970-6
phase-contrast microscopical study. Am J Trop Med 73. Murray HW. Susceptibility of Leishmania to oxygen in-
Hyg 1971;20:539-45 termediates and killing by normal macrophages. J Exp
58. Chang K-P. Leishmania donovani: promastigote-macro- Med 1981;153:1302-15
phage surface interactions in vitro. Exp Parasitol1979; 74. Nakagawara A, Nathan CF, Cohn ZA. Hydrogen perox-
48:175-89 ide metabolism in human monocytes during differen-
59. Zenian A, Rowles P, Gingell D. Scanning electron-micro- tiation in vitro. J. Clin. Invest. 1981;68:1243-52.
scopic study of the uptake of Leishmania parasites by 75. Johnson WD Jr, Mei B, Cohn ZA. The separation.Tong-
macrophages. J Cell Sci 1979;39:187-99 term cultivation, and maturation of the human mono-
60. Chang K-P. Human cutaneous leishmania in a mouse cyte. J Exp Med 1977;146:1613-26
macrophage line: propagation and isolation of intracel- 76. Murray HW, Cohn ZA. Macrophage oxygen-dependent
lular parasites. Science 1980;209:1240-2 antimicrobial activity. I. Susceptibility of Toxoplasma
61. Chang K-P. Leishmania donovan i-macrophage binding gondii to oxygen intermediates. J Exp Med 1979;150:
mediated by surface glycoproteins/antigens: character- 938-49
ization in vitro by a radioisotopic assay. Mol Biochem 77. Murray HW. Interaction of Leishmania with a macro-
Parasitol 1981;4:67-76 phage cell line: correlation between intracellular killing
62. Zenian A. Leishmania tropica: biochemical aspects of and the generation of oxygen intermediates. J Exp Med
promastigotes' attachment to macrophages in vitro. 1981;153:1690-5
Exp Parasitol 1981;51:175-87 78. Pearson RD, Steigbigel RT. Phagocytosis and killing of
63. Akiyama HJ, McQuillen NK. Interaction and transfor- the protozoan Leishmania donovani by human poly-
mation of Leishmania donovani within in vitro cul- morphonuclear leukocytes. J Immunol 1981;127: 1438-
tured cells. An electron microscopical study. Am J 43
Trop Med Hyg 1972;21:873-9 79. Buchmiiller Y, Mauel J. Studies on the mechanisms of
64. Lewis DH, Peters W. The resistance of intracellular macrophage activation: possible involvement of oxy-
Leishmania parasites to digestion by lysosomal en- gen metabolites in killing of Leishmania enriettii by ac-
zymes. Ann Trop Med Parasitol 1977;71:295-312 tivated mouse macrophages. J Reticuloendothel Soc
65. Rahman AA, Sethi KK. Intracellular behaviour of Leish- 1981;29:181-92
Immunobiology of Leishmaniasis 925

80. Avila JL, Convit J, Pinardi ME, Jacques PJ. Loss of in- leased by Leishmania donovani: immunologic relation-
fectivity of mycobacterial and protozoal exoplasmic ships with host and bacterial antigens and preliminary
parasites after exposure in vitro to the polyenzymic biochemical analysis. J Protozool 1978;25:514-25
cocktail "PIGO." Biochem Soc Trans 1976;4:680-1 99. Slutzky GM, Greenblatt CL. Isolation of a carbohydrate-
81. Reiner NE, Kazura JW. Oxidant-mediated damage of rich, immunologically active factor from cultures of
Leishmania donovani promastigotes. Infect Immun Leishmania tropica. FEBS Lett 1977;80:401-4
1982;36:1023-7 100. Dwyer DM. Antibody-induced modulation of Leishman-
82. Gottlieb M, Dwyer DM. Protozoan parasite of humans: ia donovani surface membrane antigens. J Immunol
surface membrane with externally disposed acid phos- 1976;117:2081-91
phatase. Science 1981;212:939-41 101. Lewis DH. Infection of tissue culture cells of low phago-
83. Gottlieb M, Dwyer DM. Leishmania donovani: surface cytic ability by Leishmania mexicana mexicana. Ann
membrane acid phosphatase activity of promastigotes. Trop Med Parasitol 1974;68:327-36
Exp Parasitol 1981 ;52:117-28 102. Mattock NM, Peters W. The experimental chemotherapy
84. Schmunis GA, Herman R. Characteristics of so-called of leishmaniasis. I. Techniques for the study of drug
natural antibodies in various normal sera against cul- action in tissue culture. Ann Trop Med Parasitol1975;
ture forms of Leishmania. J Parasitol 1970;56:889-96 69:349-57
85. Rezai HR, Sher S, Gettner S. Leishmania tropica, L. don- 103. Berens RL, Marr JJ. Growth of Leishmania donovani
ovani, and L. enriettii: immune rabbit serum inhibitory amastigotes in a continuous macrophage-like cell cul-
in vitro. Exp Parasitol 1969;26:257-63 ture. J Protozool 1979;26:453-6
86. Ulrich M, Ortiz DT, Convit J. The effect of fresh serum 104. Chang K-P. Leishmanicidal mechanisms of human poly-
on the leptomonads of Leishmania. I. Preliminary re- morphonuclear phagocytes. Am J Trop Med Hyg

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


port. Trans R Soc Trop Med Hyg 1968;62:825-30 1981;30:322-33
87. Hindle E, Hou PC, Patton WS. Reports from the Royal 105. Gorczynski RM, MacRae S, Kuba R, Price GB. Macro-
Society's Kala Azar Commission in China. I. Serolog- phage heterogeneity and Ir-gene control as factors in-
ical studies on Chinese kala azar. Proc R Soc Lond volved in the immune response of guinea pigs to infec-
[BioI] 1926;100:368-73 tion with Leishmania enriettii. Cell ImmunoI1981;60:
88. Adler S. Attempts to transmit visceral leishmaniasis to 367-75
man. Remarks on the histopathology of leishmaniasis. 106. Alexander J, Vickerman K. Fusion of host cell secondary
Trans R Soc Trop Med Hyg 1940;33:419-37 lysosomes with the parasitophorous vacuoles of Leish-
89. Taub J. The effect of normal human serum on Leish- mania mexicana-infected macrophages. J Protozool
mania. Bulletin of the Research Council of Israel, Sec- 1975;22:502-8
tion E 1956;6E:55-7 107. Chang K-P, Dwyer DM. Multiplication of a human para-
90. Lainson R, Strangways-Dixon J. Leishmania mexicana: site (Leishmania donovaniy in phagolysosomes of
the epidemiology of dermal leishmaniasis in British hamster macrophages in vitro. Science 1976;193:
Honduras. Trans R Soc Trop Med Hyg 1963;57:242-65 678-80
91. Ben Rachid MS. Action lytique du serum humain normal 108. Chang K-P, Dwyer DM. Leishmania donovan i-hamster
vis-a-vis de Leishmania infantum. Arch Inst Pasteur macrophage interactions in vitro: cell entry, intracellu-
Tunis 1967:44:155-61 lar survival, and multiplication of amastigotes. J Exp
92. Pearson RD, Steigbigel RT. Mechanism oflethal effect of Med 1978;147:515-30
human serum upon Leishmania donovani. J Immunol 109. Herman R. Studies of the numbers and morphology of
1980;125:2195-2201 the intracellular form of Leishmania donovani grown
93. Nussenzweig V. Reacao de fixacao do complemento para in cell culture. J Protozool 1966;13:408-18
leishmaniose visceral com anttgeno extraido do bacilo 110. Bhattacharya A, Janovy J Jr. Leishmania donovani:
da tuberculose. Tecnica, sensibilidade e especificidade. autoradiographic evidence for molecular exchanges be-
Hosptial (Rio de Janeiro) 1957;51:217-26 tween parasites and host cells. Exp Parasitol 1975;37:
94. Khaleque KA. A new method for preparing an antigen 353-60
from Kedrowsky's bacillus for the complement fixation 111. Handman E, Spira DT. Growth of Leishmania amasti-
test for kala-azar. J Pathol Bacteriol 1962;83:284-7 gotes in macrophages from normal and immune mice.
95. Camargo ME, Rebonato C. Cross-reactivity in fluores- Z Parasitenkd 1977;53:75-81
cence tests for Trypanosoma and Leishmania anti- 112. Lewis DH, Peters W. The resistance of intracellular
bodies. A simple inhibition procedure to ensure speci- Leishmania parasites to digestion by lysosomal en-
fic results. Am J Trop Med Hyg 1969;18:500-5 zymes. Ann Trop Med Parasitol 1977;71:295-312
96. Araujo FG, Mayrink W. Fluorescent antibody test in vis- 113. Berman JD, Fioretti TB, Dwyer DM. In vivo and in vitro
ceralleishmaniasis. II. Studies on the specificity of the localization of Leishmania within macrophage phago-
test. Rev Inst Med Trop Sao Paulo 1968;10:41-5 lysosomes: use of colloidal gold as a lysosomal label. J
97. Greenblatt CL, Kark JD, Schnur LF, Slutzky GM. Do Protozool 1981 ;28:239-42
Leishmania serotypes mimic human blood group anti- 114. Berman JD, Dwyer DM, Wyler DJ. Multiplication of
gens [letter]. Lancet 1981 ;1:505-6 Leishmania in human macrophages in vitro. Infect
98. Decker-Jackson JE, Honigberg BM. Glycoproteins re- Immun 1979;26:375-9
926 Pearson et al.

115. Wyler Dl. In vitro parasite-monocyte interactions in hu- sitivity, lymphocyte stimulaton, and inhibition of
man leishmaniasis. Evidence for an active role of the macrophage migration. Am 1 Trop Med Hyg 1971;20:
parasite in attachment. 1 Clin Invest 1982;70:82-8 546-51
116. Klempner MS, Wyler Dl, Andre-Schwartz 1. Plasma 131. Bryceson ADM, Bray RS, Dumonde DC. Experimental
membrane interactions between human macrophage cutaneous leishmaniasis. IV. Selective suppression of
and Leishmania tropica amastigotes [abstract]. Clin cell-mediated immunity during the response of guinea-
Res 1981;29:389A pigs to infection with Leishmania enriettii. Clin Exp
116a. Murray HW. Cell-mediated immune response in experi- Immunol 1974;16:189-201
mental visceral leishmaniasis. II. Oxygen-dependent 132. Bryceson ADM, Turk lL. The effect of prolonged treat-
killing of intracellular Leishmania donovani amasti- ment with antilymphocyte serum on the course of in-
gotes. 1 Immunol 1982:129:351-7 fections with BCG and Leishmania enriettii in the
117. Hart PD, Armstrong lA, Brown CA, Draper P. Ultra- guinea-pig. 1 PatJ101 1971;104:153-65
sturctural study of the behavior of macrophages 133. Bryceson ADM, Preston PM, Bray RS, Dumonde DC.
toward parasitic mycobacteria. Infect Immun 1972;5: Experimental cutaneous leishmaniasis. II. Effects of
803-7 immunosuppression and antigenic competition on the
118. Armstrong lA, Hart PD. Response of cultured macro- course of infection with Leishmania enriettii in the
phages to Mycobacterium tuberculosis, with observa- guinea-pig. Clin Exp Immunol 1972;10:305-35
tions on fusion of lysosomes with phagosomes. 1 Exp 134. Preston PM, Carter RL, Leuchars E, Davies A1S,
Med 1971;134:713-40 Dumonde DC. Experimental cutaneous leishmaniasis.
119. Jones TC, Hirsch lG. The interaction between Toxo- III. Effects of thymectomy on the course of infection
plasmagondii and mammalian cells. II. The absence of of CBA mice with Leishmania tropica. Clin Exp Im-

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


lysosomal fusion with phagocytic vacuoles containing munol 1972;10:337-57
living parasites. 1 Exp Med 1972;136:1173-94 135. Preston PM, Dumonde DC. Experimental cutaneous
120. Kress Y, Tanowitz H, Bloom B, Wittner M. Trypanoso- leishmaniasis. V. Protective immunity in subclinical
ma cruzi: infection of normal and activated mouse and self-healing infection in the mouse. Clin Exp Im-
macrophages. Exp Parasitol 1977;41:385-96 munol 1976;23:126-38
121. Handman E, Greenblatt CL. Promotion of leishmanial 136. Skov CB, Twohy OW. Cellular immunity to Leishmania
infections in non-permissive host macrophages by con- donovani I. The effect of T cell depletion on resistance
ditioned medium. Z Parasitenkd 1977;53:143-7 to L. donovani in mice. 1 Immunol 1974;113:2004-11
122. Slutzky GM, EI-On 1, Greenblatt CL. Leishmanial ex- 137. Skov CB, Twohy DW. Cellular immunity to Leishmania
creted factor: protein-bound and free forms from donovani. II. Evidence for synergy between thymo-
promastigote cultures of Leishmania tropica and cytes and lymph node cells in reconstitution of ac-
Leishmania donovani. Infect Immun 1979;26:916-24 quired resistance to L. donovani in mice. 1 Immunol
123. EI-On 1, Schnur LF, Greenblatt CL. Leishmania dono- 1974;113:2012-9
vani: physicochemical, immunlogical, and biological 138. Miller HC, Twohy DW. Cellular immunity to Leishmania
characterization of excreted factor from promasti- donovani in macrophages in culture. 1 Parasitol 1969;
gotes. Exp Parasitol 1979;47:254-69 55:200-7
124. BlackCDV, WatsonGl, WardRl. The use of Pent ostam 139. Smrkovski LL, Larson CL. Effect of treatment with BCG
liposomes in the chemotherapy of experimental leish- on the course of visceral leishmaniasis in BALBIc
maniasis. Trans R Soc Trop Med Hyg 1977;71:550-2 mice. Infect immun 1977;16:249-57
125. Alving CR, Steck EA, Hanson WL, Loizeaux PS, Chap- 140. Smrkovski LL, Reed SG, Larson CL. Effect of cortisone
man WL Jr, Waits VB. Improved therapy of experi- and cyclophosphmaide on the immunological role of
mental leishmaniasis by use of a liposome-encapsulated BCG in BALBIc mice challenged with Leishmania
antimonial drug. Life Sci 1978;22:1021-5 donovani. Am 1 Trop Med Hyg 1980;29:16-20
126. New RRC, Chance ML, Heath S. The treatment of ex- 141. Grimaldi GF, Moriearty PL, Hoff, R. Leishmania mexi-
perimental cutaneous leishmaniasis with liposome- cana in C3H mice: BCG and levamisole treatment of
entrapped Pentostam. Parasitology 1981;83:519-27 established infection. Clin Exp Immunol 1980;41:
127. Preston MP, Dumonde DC. Immunology of clinical and 237-42
experimental leishmaniasis. In: Cohen S, Sadun EH, 142. Mauel 1, Buchmiiller Y, Behin R. Studies on the mecha-
eds. Immunology of parasitic infections. Oxford: nisms of macrophage activation. I. Destruction of in-
Blackwell Scientific Publications, 1976 tracellular Leishmania enriettii in macrophages acti-
128. Bray RS, Bryceson ADM. Cutaneous leishmaniasis of the vated by cocultivation with stimulated lymphocytes. 1
guinea pig. Action of sensitised lymphocytes on in- Exp Med 1978;148:393-407
fected macrophages. Lancet 1968;2:898-9 143. Buchmiiller Y, MaueI 1. Studies on the mechanisms of
129. Bryceson ADM, Bray RS, Wolstencroft RA, Dumonde macrophage activation. II. Parasite destruction in mac-
DC. Immunity in cutaneous leishmaniasis of the rophages activated by supernates from concanavalin
guinea-pig. Clin Exp Immunol 1970;7:301-41 A-stimulated lymphocytes. 1 Exp Med 1979;150:
130. Blewett TM, Kadivar DMH, Soulsby ElL. Cutaneous 359-70
leishmaniasis in the guinea pig. Delayed-type hypersen- 143a. Haidaris CG, Bonventre PF. A role for oxygen-depen-
Immunobiology of Leishmaniasis 927

dent mechanisms in killing of Leishmania donovani 157. Perez H, Pocino M, Malave I. Nonspecific immuno-
tissue forms by activated macrophages. J Immunol depression and protective immunity in mice infected
1982;129:850-5 with Leishmania mexicana. Infect Immun 1981;32:
144. Nacy CA, Meltzer MS, Leonard EJ, Wyler DJ. Intracel- 415-9
lular replication and lymphokine-induced destruction 158. Scott PA, Farrell JP. Experimental cutaneous leish-
of Leishmania tropica in C3H/HeN mouse macro- maniasis. I. Nonspecific immunodepression in BALB/c
phages. J Immunol 1981;127:2381-6 mice infected with Leishmania tropica. J Immunol
145. Handman E, Burgess AW. Stimulation by granulocyte- 1981;127:2395-2400
macrophage colony-stimulating factor of Leishmania 159. Howard JG, Hale C, Liew FY. Immunological regulation
tropica killing by macrophages. J Immunol 1979;122: of experimental cutaneous leishmaniasis. IV. Prophy-
1134-7 lactic effect of sublethal irradiation as a result of abro-
146. Haidaris CG, Bonventre PF. Elimination of Leishmania gation of suppressor T cell generation in mice gene-
donovani amastigotes by activated macrophages. tically susceptible to Leishmania tropica. J Exp Med
Infect Immun 1981;33:918-26 1981;153:557-68
147. Chang K-P, Chiao JW. Cellular immunity of mice to 160. Mitchell GF, Curtis JM, Scollay RG, Handman E. Resis-
Leishmania donovani in vitro: lymphokine-mediated tance and abrogation of resistance to cutaneous leish-
killing of intracellular parasites in macrophages. Proc maniasis in reconstituted BALB/c nude mice. Aust
Nat! Acad Sci USA 1981;78:7083-7 J Exp BioI Med Sci 1981;59:539-54
148. Mauel J, Behin R, Biroum-Noerjasin, Holle B. Studies on 161. Ghose AC, Haldar JP, Pal SC, Mishra BP, Mishra KK.
protective cell-mediated mechanisms in experimental Serological investigations on Indian kala-azar. Clin
Leishmania infections. In: Van Furth R, ed. Mononu- Exp Immunol 1980;40:318-26

Downloaded from cid.oxfordjournals.org by guest on April 30, 2011


clear phagocytes in immunity, infection and pathology. 162. Biozzi G, Mouton D, Sant'Anna OA, Passos HC, Gen-
Oxford: BlackwellScientific Publications, 1975:663-72 nari M, Reis MH, Ferreira VCA, Heumann AM,
149. Berman JD, Dwyer DM. Expression of leishmania anti- Bouthillier Y, Ibanez OM, Stiffel C, Sigueira M. Gene-
gen on the surface membrane of infected human tics of immunoresponsiveness to natural antigens in the
macrophages in vitro. Clin Exp Immunol 1981;44: mouse. Curr Top Microbiol Immunol 1979;85:31-98
342-8 163. Hale C, Howard JG. Immunological regulation of experi-
150. Wyler DJ, Weinbaum FI, Herrod HR. Characterization mental cutaneous leishmaniasis. 2. Studies with Biozzi
of in vivo proliferative responses of human lympho- high and low responder lines of mice. Parasite
cytes to leishmanial antigens. J Infect Dis 1979;140: Immunol 1981;3:45-55
215-21 164. Chang K-P. Antibody-mediated inhibition of phagocyto-
151. Carvalho EM, Teixeira RS, Johnson WD Jr. Cell- sis in Leishmania donovani-human phagocyte interac-
mediated immunity in American visceral leishmaniasis: tions in vitro. Am J Trop Med Hyg 1981;30:334-9
reversible immunosuppression during acute infection. 165. Herman R. Cytophilic and opsonic antibodies in visceral
Infect Immun 1981;33:498-502 leishmaniasis in mice. Infect Immun 1980;28:585-93
152. Ghose AC, Haldar JP, Pal SC, Mishra BP, Mishra KK. 166. Desjeux P, Santoro F, Afchain D, Loyens M, Capron A.
Phytohaemagglutinin-induced lymphocyte transforma- Circulating immune complexesand anti-IgG antibodies
tion test in Indian kala-azar. Trans R Soc Trop Med in mucocutaneous leishmaniasis. Am J Trop Med Hyg
Hyg 1979;73:725-6 1980;29:195-8
153. Bryceson ADM. Diffuse cutaneous leishmaniasis in 167. Houba V, Allison AC. M-antiglobulins (rheumatoid-
Ethiopia. IV. Pathogenesis of diffuse cutaneous leish- factor-like globulins) and other gamma globulins in re-
maniasis. Trans R Soc Trop Med Hyg 1970;64:387-93 lationship to tropical parasitic infections. Lancet 1966;
154. Adler S. Immunology of leishmaniasis. Isr J Med Sci 1:848-52
1965;1:9-13 168. Carvalho EM, Andrews BS, Martinelli R, Dutra M,
155. Clinton BA, Stauber LA, Palczuk NC. Leishmania dono- Rosha M. Circulating immune complexes and rheuma-
vani:antibody response to chicken ovalbumin by in- toid factor in schistosomiasis and visceral leishmania-
fected golden hamsters. Exp Parasitol 1969;25:171-80 sis. Am J Trop Med Hyg 1983;32:61-8
156. Perez H, Arredondo B, Gonzalez M. Comparative study 169. Pearson RD, Alencar JE, Romito R, Naidu TG, Young
of American cutaneous leishmaniasis and diffuse AC, Davis JS. IV. Circulating immune complexes and
cutaneous leishmaniasis in two strains of inbred mice. rheumatoid factors in visceral leishmaniasis. J Infect
Infect Immun 1978;22:301-7 Dis 1983;147:1102

Você também pode gostar