You are on page 1of 10

Clinical and Experimental Immunology R EVI EW A RTI CL E doi:10.1111/cei.

12674

Permissive and protective roles for neutrophils in leishmaniasis

E. D. Carlsen,*† Y. Liang,† Summary


T. R. Shelite,‡ D. H. Walker,‡
Leishmania parasites are the causative agents of leishmaniasis, a neglected
P. C. Melby†‡§ and L. Soong†‡
*Department of Internal Medicine, Division of tropical disease that causes substantial morbidity and considerable mortality
Infectious Diseases, MD–PhD Combined Degree in many developing areas of the world. Recent estimates suggest that
Program, †Department of Microbiology and roughly 10 million people suffer from cutaneous leishmaniasis (CL), and
Immunology, ‡Department of Pathology, and approximately 76 000 are afflicted with visceral leishmaniasis (VL), which is
§
Department of Internal Medicine, University universally fatal without treatment. Efforts to develop therapeutics and
of Texas Medical Branch, Galveston, TX, USA vaccines have been greatly hampered by an incomplete understanding of the
parasite’s biology and a lack of clear protective correlates that must be met
in order to achieve immunity. Although parasites grow and divide
preferentially in macrophages, a number of other cell types interact with
and internalize Leishmania parasites, including monocytes, dendritic cells
and neutrophils. Neutrophils appear to be especially important shortly after
parasites are introduced into the skin, and may serve a dual protective and
permissive role during the establishment of infection. Curiously, neutrophil
recruitment to the site of infection appears to continue into the chronic
phase of disease, which may persist for many years. The immunological
impact of these cells during chronic leishmaniasis is unclear at this time. In
Accepted for publication 10 June 2015 this review we discuss the ways in which neutrophils have been observed to
Correspondence: L. Soong, Department of prevent and promote the establishment of infection, examine the role of
Microbiology and Immunology, University of anti-neutrophil antibodies in mouse models of leishmaniasis and consider
Texas Medical Branch, Medical Research recent findings that neutrophils may play a previously unrecognized role in
Building 3.132, 301 University Boulevard, influencing chronic parasite persistence.
Galveston, TX 77555-1070, USA.
E-mail: lysoong@utmb.edu Keywords: Leishmania, monoclonal antibody, mouse model, neutrophil

Introduction fission in macrophages before escaping into the extracellular


Leishmania spp. are a group of obligately intracellular proto- space and seeking new host phagocytes. The parasite’s life
zoan parasites belonging to the Trypanosomitida order. cycle is completed when sandflies consume blood containing
Leishmania parasites are distributed widely in the tropics and parasitized cells. Subsequently, sandfly-ingested amastigotes
subtropics and are the causative agents of a cluster of clinical rapidly convert back into promastigotes.
diseases known as leishmaniases. Leishmania parasites alter- In humans, symptomatic leishmaniasis is highly variable
nate between two life-cycle stages that are highly adapted to in its clinical presentation due to differences in the infect-
the distinct environments of the parasite’s life cycle. Flagel- ing Leishmania species and the patient’s immune status.
lated promastigotes reside in the midgut of infected female Most Leishmania species that cause illness in humans ini-
sandflies and gain access to the skin of vertebrate hosts when tially induce a disease manifestation known as localized
sandflies take a bloodmeal. In the skin, promastigotes are cutaneous leishmaniasis (CL), which is characterized by
internalized rapidly by residential and recruited cell popula- singular or multiple well-demarcated ulcerations of the
tions, with macrophages being the parasite’s primary target skin that correspond to sandfly bite sites. The time between
cells. Within macrophages, promastigotes convert into amas- parasite inoculation and the development of ulcerated
tigotes, which actively replicate and cause disease in mam- lesions is highly variable, but typically takes weeks to
malian hosts. Amastigotes undergo several rounds of binary months [1]. In many cases, localized CL is self-limiting

C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V 109
E. D. Carlsen et al.

after patients develop protective adaptive immunity, Although neutrophils are frequently the first-responders
although extensive scarification at the site of resolved during acute infection or tissue damage, it is important to
lesions is typical. However, lesion resolution may be hin- consider that these cells are also encountered commonly in
dered in immunocompromised patients and in those chronic inflammatory and infectious conditions, including
infected with particular parasite species (such as those rheumatoid arthritis [33], systemic lupus erythematosis
belonging to the L. mexicana complex) [2]. It is important [34], leprosy [35] and tuberculosis [36]. Recent studies
to emphasize that in the absence of pharmacotherapy, pro- have also observed neutrophils in the chronic lesions of
tective immunity and lesion resolution are not synony- animals that have been infected naturally or experimentally
mous with sterile cure (i.e. the healing of ulcers does not with Leishmania [37–39], and several reports have identi-
correlate with complete parasite elimination from the site fied neutrophils as a prevalent component of the chronic
of infection) [3]. Therefore, factors that alter host immune inflammatory infiltrate in patients with CL [40–42], diffuse
status (such as corticosteroids or HIV) can potentially trig- cutaneous leishmaniasis [43] and mucocutaneous leishma-
ger disease reactivation in clinically resolved patients [4,5]. niasis [44]. In this review, we will discuss the role of neu-
Additionally, resolution of localized CL does not preclude trophils in acute and chronic leishmaniasis, with a
patients from experiencing several secondary forms of particular focus on neutrophil–parasite interactions and
leishmaniasis, including diffuse cutaneous leishmaniasis anti-neutrophil antibody treatments in experimental mod-
and mucocutaneous leishmaniasis. These sequelae are par- els of leishmaniasis.
ticularly alarming because they can be severely disfiguring.
For more information pertaining to these more rare forms
Direct neutrophil–promastigote interactions
of disease, readers are directed to an excellent clinical
review of human leishmaniasis in its many forms [1]. Several elegant studies have demonstrated the rapid
Of note, Leishmania species in the L. donovani complex recruitment of neutrophils to the site of promastigote
(including L. donovani, L. infantum and L. chagasi) cause infection in the skin [9,45–47]. However, the role of neu-
little to no pathology upon entering the skin, but instead trophils during the early stages of infection is complex and
disseminate to the bone marrow, spleen and liver to cause multi-faceted. We and others have demonstrated that neu-
visceral leishmaniasis (VL) [6]. VL is the most severe form trophils possess some direct leishmanicidal activity in vitro,
of leishmaniasis, characterized by fever, cachexia, hepatos- suggesting that these cells may represent an important
plenomegaly and pancytopenia. In the absence of medical component of the early immune response against incoming
management, VL is fatal due to secondary infection and/or promastigotes [48,49]. However, as evidenced by parasite
coagulopathy [7,8]. Patients who survive VL are at risk for persistence at the site of infection, direct neutrophil killing
a unique form of secondary leishmaniasis known as post- of promastigotes is clearly insufficient in controlling the
kala-azar cutaneous leishmaniasis [1]. establishment of infection and the development of clinical
Because neutrophils recruit rapidly to the site of infec- disease.
tion and engage in early contact with Leishmania promasti- Reports that a subset of promastigotes can survive for
gotes [9,10], these cells (and their role in early parasite prolonged periods in neutrophils without sustaining lethal
recognition and clearance) have been the subject of active damage also highlight additional, non-microbicidal roles
research in recent years [11–22]. Neutrophils play a critical for these cells during Leishmania infection [14,15,17]. Neu-
role as first-line defenders against invading microbes and trophils that contact (but do not kill) promastigotes may
utilize a number of innate techniques to aid in pathogen act as ‘Trojan horses’ or ‘Trojan rabbits’ [50], whereby par-
recognition and clearance. Importantly, they can exert asitized cells or free parasites that have escaped from neu-
direct microbicidal activity in the form of proteolytic trophils, respectively, can be internalized readily by
enzymes [23,24], reactive oxygen species [25] and neutro- neighbouring phagocytes (such as macrophages or dendri-
phil extracellular traps [26]. In addition, neutrophils can tic cells) in a manner that enhances parasite infectivity and
secrete a wide array of cytokines and chemokines, which persistence [9,18,51]. Therefore, it is likely that neutrophils
helps in the recruitment and education of more specialized play a dual protective and permissive role shortly after pro-
cells to the site of insult [27]. Even upon dying, neutrophils mastigote infection by reducing incoming parasite burden
can impact upon their surroundings to encourage the and subsequently facilitating safe passage of surviving para-
amplification of inflammatory cascades via necrosis [28], sites to naive host cells.
or facilitate immune response resolution and wound heal- Curiously, neutrophil recruitment and function at the site
ing via apoptosis [29]. Finally, roles for neutrophils in anti- of experimental infection may also differ depending on the
gen presentation [30], regulating T cell proliferation [31] site and route of parasite inoculation. Ribeiro-Gomes et al.
and driving B cell class-switching and antibody production recently compared neutrophil recruitment and early parasite
[32] have also been identified, suggesting that neutrophils clearance in C57BL/6 mice that received L. major promasti-
may exert far more diverse functions than suspected gotes in the pinnae intradermally (i.d.) or in the footpad sub-
initially. cutaneously (s.c.) [52]. Mice receiving intradermal parasites

110 C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V
Complex roles for neutrophils in leishmaniasis

exhibited a greater influx of neutrophils to the site of infec- C57BL/6 mice or macrophages revealed that neutrophils
tion, and these cells were better able to internalize promasti- could facilitate parasite clearance [61]. However, host back-
gotes when compared to mice infected subcutaneously. This ground may be less important with regard to New World
early neutrophilic response corresponded with a 10-fold Leishmania infections, as neutrophils enhanced the ability
higher parasite burden in intradermally infected mice versus of macrophages to kill L. amazonensis [62] and L. brazilien-
subcutaneously infected mice, and underscores the potential sis [63] in vitro, regardless of which mouse strain was used.
for distinct immunological outcomes when comparing infec-
tion in different tissue compartments [52]. Comparing
Neutrophils in visceral leishmaniasis
L. major infection via needle inoculation and natural insect
vector has also yielded interesting results related to neutro- VL-induced neutropenia has been a clinically described
phil function. Specifically, Peters et al. demonstrated that entity for decades [66], and appears to be a risk factor for
neutrophil infiltration was more prolonged in response to death due to secondary infection in VL patients [67]. It has
L. major promastigotes administered via natural sandfly bite long been proposed that neutropenia is probably related
[53], and neutrophil depletion in this model improved para- to increased granulocyte clearance in the spleen [68],
site clearance and amplified the immune response generated although more recent reports suggest that anti-neutrophil
by an experimental killed vaccine [9,53]. antibodies may also play a role [69]. Interestingly, a small
clinical trial demonstrated that the addition of granulo-
cyte–macrophage colony-stimulating factor (GM-CSF)
Impact of neutrophils upon macrophage infection
to standard pentavalent antimonial therapy improved neu-
In addition to their role in direct pathogen clearance, neu- tropenia in VL patients and reduced their risk of acquiring
trophils can also influence the local inflammatory environ- secondary infection. However, adding GM-CSF failed to
ment and the development of protective immunity by directly accelerate the resolution of VL [70]. Almeida and
interacting with a number of immune cell types, such as colleagues reported recently that derangements in neutro-
dendritic cells, macrophages, natural killer (NK) cells, T phil numbers and function also appear to be important
cells and B cells [32,54–59]. Because of the important role characteristics of canine VL [71]. In dogs with VL, the neu-
of macrophages during Leishmania infection, several trophil phenotype appears to vary depending on the sever-
groups have investigated the cross-talk between neutrophils ity of disease. Neutrophils from severely affected dogs
and infected macrophages [60–64]. Collectively, these stud- undergo oxidative burst poorly and are more prone to apo-
ies indicate that neutrophils can either promote parasite ptosis. In contrast, neutrophils from dogs with less severe
clearance or facilitate parasite growth and survival in mac- disease undergo oxidative burst more readily and appear to
rophages, depending on several critical factors. Afonso have improved half-life [71].
et al. reported that apoptotic and necrotic neutrophils had
opposing effects on L. amazonensis promastigote survival
Anti-neutrophil treatment in mouse models
and growth in human macrophages [60]. Neutrophils that
had undergone apoptosis in response to ultraviolet (UV) Anti-neutrophil antibody treatment is a popular method
irradiation strongly promoted parasite growth in macro- for assessing the role of neutrophils in various infectious
phages in a transforming growth factor (TGF)-b1 and and inflammatory diseases in vivo [72–75]. A handful of
prostaglandin E2 (PGE2)-dependent manner. In contrast, studies have utilized this approach to determine the role of
freeze/thawed necrotic neutrophils facilitated parasite clear- neutrophils after Leishmania promastigote infection, but
ance in macrophages via a tumour necrosis factor (TNF)-a conflicting results and considerable differences in experi-
and neutrophil elastase-dependent mechanism. Surpris- mental design between studies make it difficult to draw
ingly, live neutrophils had little effect on the percentage of concrete conclusions (Table 1). For example, Chen et al.
infected macrophages or total parasite burden [60]. and Tacchini-Cottier et al. examined the effects of neutro-
Historically, mouse strain-specific differences in suscep- phil depletion on L. major burden and lesion progression
tibility to L. major have been attributed largely to differen- in BALB/c mice, but reported contradictory findings
ces in T helper cell polarization, with susceptible BALB/c [12,20]. It is possible that the use of different parasite
mice preferentially adopting a T helper type 2 (Th2) strains and dissimilar neutrophil depletion antibodies/dos-
response and resistant C57BL/6 mice adopting a Th1-type ing schedules contributed partially to the discrepancies in
response [65]. More recently, Ribeiro-Gomes et al. demon- these studies. Notably, contrary observations have also
strated that the anti-parasite responses of these two strains been published regarding the role of neutrophils in experi-
of mice also differ at the innate immune level [61]. In mental VL. Several studies reported that anti-neutrophil
L. major-infected BALB/c mice or BALB/c-derived macro- treatment during L. donovani and L. infantum infection
phages, adding syngeneic exudate neutrophils (whether exacerbated visceralization [16,77,78], while Thalhofer
intact, necrotic or apoptotic) increased parasite growth et al. noted that antibody treatment reduced parasite
substantially. In contrast, equivalent experiments in carriage transiently in the draining lymph nodes of

C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V 111
112
Table 1. Anti-neutrophil treatment in mouse models of cutaneous and visceral leishmaniasis. Studies utilizing anti-neutrophil antibodies in mouse models of Leishmania major, L. donovani, L. infan-
tum, L. braziliensis and L. amazonensis infection suggest that these cells can play an important and complex role in the anti-parasite immune response and can impact disease progression.
Parasite species/strain Antibody treatment Host strain Effects of antibody treatment
E. D. Carlsen et al.

L. major LV39 NIMP-R14 1 mg 26 h BALB/c Delayed lesion progression, reduced parasite burden
C57BL/6 Accelerated early lesion progression, time required for lesion resolution
unaffected, no differences in parasite burden [20]
L. major 5ASKH RB6-8C5 250 lg Day 21 BALB/c Accelerated lesion progression and enhanced parasite burden
C3H/HeJ No change in lesion progression [12]
L. major Bokkara RB6-8C5 2 mg Days 23, 0, 3 BALB/c Accelerated lesion progression between weeks 1 and 6, chronic progression
unaltered
C57BL/6 Accelerated lesion progression at weeks 3 and 4, higher dLN burden at days

V
16 and 29, chronic progression unaltered [76]
L. major Friedlin V1 (sandfly) NIMP-R14 0.5 mg Days 3.5, 9, and 14 C57BL/6 Enhanced parasite clearance and stronger IFN-g and TNF-a responses
induced by a killed Leishmania vaccine in neutrophil-depleted animals
[53]
L. major Friedlin V1 RB6-8C5 500 lg 216 h C57BL/6 Depletion prior to sandfly-mediated infection reduced parasite burden at
the site of infection at 1 and 4 weeks [9]
L. donovani LV82 NIMP-R14 250 lg Days 0, 3, 6, 9, 12 BALB/c Increased burden in the spleen and bone marrow, hepatosplenomegaly [16]
L. donovani LV9 RB6-8C5 200 lg Days 21, 2, 5, 8, 11, 14 BALB/c Enhanced parasite burden in the liver and spleen
C57BL/6 Enhanced parasite burden in the liver and spleen [77]
L. infantum MON1 RB6-8C5 200 lg 248 h, 25 h, 72 h, 168 h BALB/c Increased parasite burden in the spleen, but not the liver
RB6-8C5 200 lg starting at 3 months, No effect [78]
2 doses/week for 4 weeks
L. chagasi MHOM/BR/00/1669 RB6-8C5 25 lg Day 21 BALB/c Impaired extracellular parasite clearance and reduced parasite carriage in
draining lymph node cells on day 1. At day 3 and onwards, parasite bur-
den was similar in control and antibody-treated animals.
1A8 25 lg Day 21 Impaired extracellular parasite clearance and reduced parasite carriage in
draining lymph node cells on day 1 [47]
L. braziliensis BA788 RB6-8C5 500 lg Days 21, 3, 6, 9, 12 BALB/c Increased parasite burden at the site of infection and in the dLNs at 2
weeks, increased IL-2, IL-4, IL-5, IFN-g, TNF-a, faster early lesion pro-
gression [63]
L. amazonensis PH8 RB6-8C5 500 lg 216 h BALB/c Increased lesion size and parasite burden at 1 week
C57BL/6 No effect
RB6-8C5 500 lg Days 21, 1, 3, 5, 7 BALB/c Increased lesion size at weeks 4 and 8
C57BL/6 No effect [79]
L. mexicana MYNC/BZ/62/M379 1A8 150 lg 26 h, 36 h C57BL/6 Decreased carriage of parasites in phagocytes and lesion resolution [80]
IL 5 interleukin; IFN 5 interferon; TNF 5 tumour necrosis factor.

C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
Complex roles for neutrophils in leishmaniasis

L. chagasi-infected mice at day 1 post-inoculation without activation and microbicidal activity following infection
impacting the parasite burden at day 3 [47]. with L. amazonensis [48], a species responsible for diffuse
Although the effects of anti-neutrophil treatment vary CL in humans [86] and non-healing CL in mice [2]. We
greatly in these published reports, most of them indicate found that murine neutrophils internalized L. amazonensis
that neutrophils can play some sort of a protective role promastigotes and amastigotes efficiently after 4 h, and
shortly after parasite infection. However, several studies both parasite forms triggered neutrophil activation, oxida-
highlight an opposing regulatory role for neutrophils, tive burst and early neutrophil death. However, amastigotes
implying that these cells may possess considerably more and promastigotes were very different in their susceptibility
plasticity than appreciated previously. Regardless, these to neutrophil microbicidal mechanisms. Notably, while
studies demonstrate co-operatively that neutrophils are more than half of promastigotes were cleared by cells
involved in the early anti-parasite response, and suggest within 6 h, nearly 100% of amastigotes survived prolonged
that disrupting normal neutrophil functions at the site of co-culture with neutrophils [48]. These findings corrobo-
infection can influence parasite growth and disease pathol- rated and expanded upon our previous observations that
ogy potently. L. amazonensis amastigotes were highly resistant to purified
One possible confounding factor in the previous reports is human histone proteins, which are an important anti-
the anti-neutrophil antibodies utilized. Many studies used microbial component of neutrophil extracellular traps
RB6-8C5, but in recent years the specificity of this clone for [22]. Additionally, the consensus findings from these
neutrophils has been questioned [19]. Importantly, RB6-8C5 in-vitro studies highlight the superior resistance of L. ama-
also appears to target non-neutrophil myeloid cell popula- zonensis amastigotes against neutrophil microbicidal
tions, including some plasmacytoid dendritic cells [81], defence, even in the face of amastigote-mediated activation
monocytes [82], eosinophils [83] and T lymphocytes [84]. and oxidative burst [48].
Additionally, the findings of a compensatory increase in neu- In an effort to understand neutrophil interactions with
trophil numbers above the controls in the footpad [79] and different Leishmania species more clearly, we recently
blood [76,78] of Leishmania-infected mice after completing assessed neutrophil activation, cytokine production and
RB6-8C5 treatments make it difficult to determine whether microbicidal activity in response to amastigotes of L. brazil-
the observed phenotypes were due to initial neutrophil deple- iensis, the species responsible for mucocutaneous leishmani-
tion or subsequent neutrophil influx. With the advent of asis in humans [87] and self-healing CL in mice [88].
newer anti-neutrophil antibody clones (such as 1A8), it has We were surprised to find that amastigotes of L. amazonensis
become possible to target these cells with greater specificity. and L. braziliensis triggered distinct neutrophil responses.
Interestingly, Wang et al. demonstrated that very low doses of L. braziliensis amastigotes were a far stronger stimulus for
1A8 (as little as 5 lg per animal) can block neutrophil recruit- neutrophil activation, oxidative burst, degranulation and
ment to sites of inflammation without inducing protracted proinflammatory cytokine production than L. amazonensis
periods of systemic neutropenia [75], which may help to pre- amastigotes [64]. Additionally, neutrophils exerted measura-
serve unrelated neutrophil functions at distal sites, such as ble leishmanicidal activity against L. braziliensis amastigotes,
immunoglobulin production in the spleen [32]. Therefore, while L. amazonensis amastigotes were highly resistant to
additional studies to investigate the impact of low-dose 1A8 neutrophil microbicidal mechanisms [64]. These in-vitro
treatment on promastigote infection will improve our under- findings support the notion that these two Leishmania spe-
standing of neutrophils in acute leishmaniasis. cies differ in their ability to trigger (or evade) immune rec-
ognition upon entering a host [89,90] and emphasize that
neutrophils may interact with amastigotes from different
Amastigote–neutrophil interactions: implications
Leishmania species in vastly different ways. Currently, it is
for chronic cutaneous leishmaniasis
unclear whether neutrophils also respond to L. amazonensis
Despite clear evidence of sustained neutrophil recruitment and L. braziliensis amastigotes differently in vivo and how
during chronic leishmaniasis [37–44], and the finding that such differential responses impact on the chronic phase of
reduced neutrophil recruitment correlates with decreased leishmaniasis.
lesion ulceration in infected mice [85], the contribution of Reports identifying neutrophils in the chronic lesions of
these cells to amastigote clearance and the maintenance patients [42–44] and animals [37–39,91] prompted us to
of a low-grade inflammatory response and tissue damage analyse these cells in detail using a persistent model of
is incompletely understood. Currently, there is a scarcity L. amazonensis infection. We found that C57BL/6 mice
of studies examining neutrophil–amastigote interactions infected with L. amazonensis for as long as 14 weeks had
in vitro, and anti-neutrophil studies assessing the impact notable accumulations of neutrophils in infected, non-
of these cells during the chronic phase of cutaneous leish- ulcerated footpads (Fig. 1a), indicating that neutrophil
maniasis are largely absent from the literature. To deter- recruitment to the site of infection persists during chronic
mine whether neutrophils respond to amastigotes and disease. Additionally, we detected a small but appreciable
promastigotes differently, we recently compared neutrophil population (0.2%) of lymphocyte antigen 6 complex G

C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V 113
E. D. Carlsen et al.

Fig. 1. Neutrophil detection in the tissues of a


mouse model of chronic cutaneous leishmaniasis.
(a) At 14 weeks of infection with Leishmania
amazonensis, frozen footpad sections from
C57BL/6 mice were stained for myeloperoxidase
(MPO, red). Shown is a representative section
containing MPO-positive cells with distinct
polymorphonuclear phenotype. (b) Flow
cytometric detection of Ly6C1Ly6G1 neutrophils
in the draining lymph nodes of similarly treated
mice.

locus/lymphocyte antigen 6 complex C locus (Ly6G1/ utilize the 1A8 clone, it was unclear whether 1A8 treatment
Ly6C1) neutrophils in the draining lymph nodes of chroni- resulted in neutrophil depletion or whether Ly6G was sim-
cally infected mice (Fig. 1b). To assess the impact of neutro- ply blocked on circulating cells. To address this issue, we
phils during the chronic stage of L. amazonensis infection developed a Ly6G-independent flow cytometry panel, in
without altering early host–parasite interactions, we treated which cells were co-stained for myeloperoxidase (MPO, a
preinfected mice with anti-neutrophil antibodies. To marker highly expressed in neutrophils, but also in rela-
achieve this, mice were infected with 2 3 105 metacyclic tively low levels in macrophages and monocytes [92,93])
promastigotes and infections were allowed to proceed until and 7/4 (a marker for macrophages, monocytes, neutro-
the onset of visible footpad swelling at 4 weeks. Subse- phils and myeloid progenitor cells [94]). We found that
quently, mice received intraperitoneal injections of 300 lg co-staining for MPO and 7/4 could be used in lieu of Ly6G
of anti-Ly6G (1A8) or isotype control (2A3) every 6 days. staining to identify the neutrophil population in circulating
Repeated 1A8 treatment efficiently blocked Ly6G on circu- CD11b1 cells, and that MPOhi 7/41 cells were equivalent to
lating neutrophils, as assessed by reduced binding of allo- Ly6G1 cells (Fig. 2b). This panel revealed that repeat anti-
phycocyanin (APC)-conjugated anti-Ly6G (Fig. 2a). As our neutrophil treatment did not substantially alter the fre-
treatment antibody and APC-conjugated antibody both quency of CD11b1MPOhi 7/41 neutrophils in the blood at

Fig. 2. Effect of anti-neutrophil treatment on


circulating neutrophils and impact of treatment
on progression of chronic leishmaniasis. Four
weeks post-infection with Leishmania
amazonensis promastigotes, C57BL/6 mice were
treated with 1A8 antibody or 2A3 isotype control
at 6-day intervals for a total of 6 weeks. (a) Flow
cytometry of blood samples collected at 6 weeks
post-antibody treatment, indicating efficient
binding of lymphocyte antigen 6 complex G
locus (Ly6G) in circulating leucocytes. (b)
Surrogate flow cytometry panel utilizing
myeloperoxidase (MPO) and 7/4 to identify
neutrophils in lieu of Ly6G staining. Ly6G– cells
are shown in grey, while Ly6G1 cells are shown
in blue. (c) Comparison of neutrophil
percentages in circulating CD11b1 leucocytes in
response to serial antibody treatments. (d) Effect
of repeated antibody treatment on lesion
progression in preinfected mice (10 animals were
included in each antibody treatment group).
Two-way analysis of variance (ANOVA) indicates
that the progressions are statistically different.
**P < 001 and ***P < 0001 indicate statistically
significant differences between groups at
designated time-points using the Bonferroni
method. Data shown are from one of two
conducted experiments with similar trends.

114 C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V
Complex roles for neutrophils in leishmaniasis

the time of killing (Fig. 2c). Nevertheless, mice receiving implications on the outcome of infection. In order to
prolonged anti-neutrophil treatment (between 4 and 10 assess reliably the impact of neutrophils in the context of
weeks post-infection) exhibited lesions with a significantly infection in the future, it will be helpful for researchers
greater rate of growth than mice receiving isotype control to reach a consensus regarding the use of prototype para-
(Fig. 2d). Surprisingly, anti-neutrophil treatment did little site strains, highly neutrophil-specific antibodies and sim-
to alter parasite burden in infected footpads (data not ilar dosing regimens that minimize compensatory and/or
shown), suggesting that neutrophils may limit detrimental off-target effects.
tissue damage during chronic disease without exerting Importantly, despite decades of research, there is no
extensive leishmanicidal activity during L. amazonensis anti-Leishmania vaccine approved for human use, which is
infection. The results of this study identify neutrophils as a due partially to the fact that clear correlates of immune
component of the chronic inflammatory infiltrate at the protection are lacking for certain parasite species. For
site of infection and in the draining lymph nodes, and sug- example, although a strong Th1 response is sufficient for
gest that neutrophils may impact the inflammatory milieu control of L. major [95,96], effector Th1 cells are patho-
to modulate lesion progression. While exciting, the results genic during experimental L. amazonensis infection [97],
of this initial study highlight the necessity of investigating and stimulating L. amazonensis-infected macrophages with
the detailed in-vivo function of neutrophils during chronic interferon (IFN)-g can actually accelerate parasite growth
leishmaniasis, as well as the contribution of these cells to [98]. Consequently, the development of future vaccines
the pathophysiology of other chronic infectious and inflam- and therapeutics for leishmaniasis may benefit greatly from
matory disorders. unconventional strategies that focus on amplifying innate
immune functions (particularly those functions that can
Animal care and use exert direct leishmanicidal activity). We suspect that
All animals were maintained under specific pathogen-free obtaining a clearer understanding of how neutrophils con-
conditions and used in accordance with protocols approved tribute to anti-parasite immunity and inflammation will be
by the Animal Care and Use Committee of the University critical to this tactic.
of Texas Medical Branch (Galveston, TX, USA).
Acknowledgements
Conclusions The authors would like to thank Dr Hui Wang for technical
immunohistochemistry assistance and Victor Hugo Carpio
Animal models of leishmaniasis indicate that neutrophils
for helpful flow cytometry suggestions. The authors also
are a prominent component of the inflammatory infiltrate
wish to express gratitude to other members of the UTMB
throughout the course of parasite infection, and several
Joint Immunology Working Group (Drs Y. Cong, R. Ste-
clinical reports confirm that neutrophils are also present in
phens, R. Rajsbaum, J. Sun and their trainees) for helpful
the persistent lesions of human patients. Despite recent
discussions. This work was supported in part by NIAID
suggestions that neutrophils may play an important role in
grants (R56 AI043003; R03 AI103812) to L.S., NIAID grant
maintaining the inflammatory environment in patients
(R21 AI107419) to P.M. and the James W. McLaughlin
with cutaneous and mucocutaneous leishmaniasis [40,44],
Predoctoral Fellowship and Blocker Scholar Fellowship in
few studies have investigated the role of these cells in the
Biomedical Research to E.C.
chronic disease process. Mounting evidence suggests that
neutrophils are not passive bystanders in disease pathoge-
nesis and immunopathology; rather, it is probable that Disclosure
neutrophils play a complex and active role in parasite rec-
The authors declare no competing interests.
ognition and the anti-parasite response in both acute and
chronic leishmaniasis.
References
In-vivo anti-neutrophil treatment is a powerful method
for ascertaining the role of neutrophils in animal models 1 Paniz Mondolfi AE, Duffey GB, Horton LE et al. Intermediate/
of infectious and inflammatory diseases, especially when borderline disseminated cutaneous leishmaniasis. Int J Dermatol
2013; 52:446–55.
focusing on their function during a specific phase or
2 Soong L. Subversion and utilization of host innate defense by
period of disease progression. However, poor neutrophil
Leishmania amazonensis. Front Immunol 2012; 3:58.
specificity and observations of compensatory neutrophilia
3 Mendonca MG, de Brito ME, Rodrigues EH, Bandeira V,
make past murine leishmaniasis studies difficult to inter- Jardim ML, Abath FG. Persistence of Leishmania parasites in
pret. Despite these limitations, recent studies help to scars after clinical cure of American cutaneous leishmaniasis: is
establish that neutrophils are important regulators of the there a sterile cure? J Infect Dis 2004; 189:1018–23.
anti-Leishmania immune response and aid in demon- 4 Tuon FF, Sabbaga Amato V, Floeter-Winter LM et al. Cutaneous
strating that the presence of these cells has important leishmaniasis reactivation 2 years after treatment caused by

C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V 115
E. D. Carlsen et al.

systemic corticosteroids – first report. Int J Dermatol 2007; 46: 23 Massberg S, Grahl L, von Bruehl ML et al. Reciprocal coupling
628–30. of coagulation and innate immunity via neutrophil serine pro-
5 Okwor I, Uzonna JE. The immunology of Leishmania/HIV co- teases. Nat Med 2010; 16:887–96.
infection. Immunol Res 2013; 56:163–71. 24 Hahn I, Klaus A, Janze AK et al. Cathepsin G and neutrophil
6 McGwire BS, Satoskar AR. Leishmaniasis: clinical syndromes elastase play critical and nonredundant roles in lung-protective
and treatment. Q J Med 2014; 107:7–14. immunity against Streptococcus pneumoniae in mice. Infect
7 Belo VS, Werneck GL, Barbosa DS et al. Factors associated with Immun 2011; 79:4893–901.
visceral leishmaniasis in the Americas: a systematic review and 25 Bonfim-Mendonca Pde S, Ratti BA, Godoy Jda S et al. b-glucan
meta-analysis. PLOS Negl Trop Dis 2013; 7:e2182. induces reactive oxygen species production in human neutro-
8 Werneck GL, Batista MS, Gomes JR, Costa DL, Costa CH. Prog- phils to improve the killing of Candida albicans and Candida
nostic factors for death from visceral leishmaniasis in Teresina, glabrata isolates from vulvovaginal candidiasis. PLOS ONE
Brazil. Infection 2003; 31:174–7. 2014; 9:e107805.
9 Peters NC, Egen JG, Secundino N et al. In vivo imaging reveals 26 Lu T, Kobayashi SD, Quinn MT, Deleo FR. A NET Outcome.
an essential role for neutrophils in leishmaniasis transmitted by Front Immunol 2012; 3:365.
sand flies. Science 2008; 321:970–4. 27 Tecchio C, Micheletti A, Cassatella MA. Neutrophil-derived
10 Laskay T, van Zandbergen G, Solbach W. Neutrophil granulo- cytokines: facts beyond expression. Front Immunol 2014; 5:508.
cytes – Trojan horses for Leishmania major and other intracellu- 28 Heijink IH, Pouwels SD, Leijendekker C et al. Cigarette Smoke
lar microbes? Trends Microbiol 2003; 11:210–4. Induced DAMP Release from Necrotic Neutrophils Triggers
11 Charmoy M, Megnekou R, Allenbach C et al. Leishmania major Pro-inflammatory Mediator Release. Am J Respir Cell Mol Biol
induces distinct neutrophil phenotypes in mice that are resistant 2015; 52:554–62.
or susceptible to infection. J Leukoc Biol 2007; 82:288–99. 29 El Kebir D, Gjorstrup P, Filep JG. Resolvin E1 promotes
12 Chen L, Zhang ZH, Watanabe T et al. The involvement of neu- phagocytosis-induced neutrophil apoptosis and accelerates reso-
trophils in the resistance to Leishmania major infection in sus- lution of pulmonary inflammation. Proc Natl Acad Sci USA
ceptible but not in resistant mice. Parasitol Int 2005; 54:109–18. 2012; 109:14983–8.
13 Gabriel C, McMaster WR, Girard D, Descoteaux A. Leishmania 30 Kambayashi T, Laufer TM. Atypical MHC class II-expressing
donovani promastigotes evade the antimicrobial activity of neu- antigen-presenting cells: can anything replace a dendritic cell?
trophil extracellular traps. J Immunol 2010; 185:4319–27. Nat Rev Immunol 2014; 14:719–30.
14 Gueirard P, Laplante A, Rondeau C, Milon G, Desjardins M. 31 Hock BD, Taylor KG, Cross NB, Kettle AJ, Hampton MB,
Trafficking of Leishmania donovani promastigotes in non-lytic McKenzie JL. Effect of activated human polymorphonuclear leu-
compartments in neutrophils enables the subsequent transfer of cocytes on T lymphocyte proliferation and viability. Immunol-
parasites to macrophages. Cell Microbiol 2008; 10:100–11. ogy 2012; 137:249–58.
15 Laufs H, Muller K, Fleischer J et al. Intracellular survival of 32 Puga I, Cols M, Barra CM et al. B cell-helper neutrophils stimu-
Leishmania major in neutrophil granulocytes after uptake in the late the diversification and production of immunoglobulin in
absence of heat-labile serum factors. Infect Immun 2002; 70: the marginal zone of the spleen. Nat Immunol 2012; 13:170–80.
826–35. 33 Verri WA Jr, Souto FO, Vieira SM et al. IL-33 induces neutro-
16 McFarlane E, Perez C, Charmoy M et al. Neutrophils contribute phil migration in rheumatoid arthritis and is a target of anti-
to development of a protective immune response during onset TNF therapy. Ann Rheum Dis 2010; 69:1697–703.
of infection with Leishmania donovani. Infect Immun 2008; 76: 34 Leffler J, Martin M, Gullstrand B et al. Neutrophil extracellular
532–41. traps that are not degraded in systemic lupus erythematosus
17 Mollinedo F, Janssen H, de la Iglesia-Vicente J, Villa-Pulgarin activate complement exacerbating the disease. J Immunol 2012;
JA, Calafat J. Selective fusion of azurophilic granules with Leish- 188:3522–31.
mania-containing phagosomes in human neutrophils. J Biol 35 Lee DJ, Li H, Ochoa MT et al. Integrated pathways for neutro-
Chem 2010; 285:34528–36. phil recruitment and inflammation in leprosy. J Infect Dis 2010;
18 Ribeiro-Gomes FL, Peters NC, Debrabant A, Sacks DL. Efficient 201:558–69.
capture of infected neutrophils by dendritic cells in the skin 36 Lowe DM, Redford PS, Wilkinson RJ, O’Garra A, Martineau
inhibits the early anti-Leishmania response. PLOS Pathog 2012; AR. Neutrophils in tuberculosis: friend or foe? Trends Immunol
8:e1002536. 2012; 33:14–25.
19 Ribeiro-Gomes FL, Sacks D. The influence of early neutrophil- 37 Lopez Kostka S, Dinges S, Griewank K, Iwakura Y, Udey MC,
Leishmania interactions on the host immune response to infec- von Stebut E. IL-17 promotes progression of cutaneous leishma-
tion. Front Cell Infect Microbiol 2012; 2:59. niasis in susceptible mice. J Immunol 2009; 182:3039–46.
20 Tacchini-Cottier F, Zweifel C, Belkaid Y et al. An immunomo- 38 Souza-Lemos C, de-Campos SN, Teva A et al. Dynamics of
dulatory function for neutrophils during the induction of a immune granuloma formation in a Leishmania braziliensis-
CD41 Th2 response in BALB/c mice infected with Leishmania induced self-limiting cutaneous infection in the primate Macaca
major. J Immunol 2000; 165:2628–36. mulatta. J Pathol 2008; 216:375–86.
21 Xin L, Vargas-Inchaustegui DA, Raimer SS et al. Type I IFN 39 Vercosa BL, Melo MN, Puerto HL, Mendonca IL, Vasconcelos
receptor regulates neutrophil functions and innate immunity to AC. Apoptosis, inflammatory response and parasite load in skin
Leishmania parasites. J Immunol 2010; 184:7047–56. of Leishmania (Leishmania) chagasi naturally infected dogs: a
22 Wang Y, Chen Y, Xin L et al. Differential microbicidal effects of histomorphometric analysis. Vet Parasitol 2012; 189:162–70.
human histone proteins H2A and H2B on Leishmania promasti- 40 Morgado FN, Schubach A, Rosalino CM et al. Is the in situ
gotes and amastigotes. Infect Immun 2011; 79:1124–33. inflammatory reaction an important tool to understand the

116 C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V
Complex roles for neutrophils in leishmaniasis

cellular immune response in American tegumentary leishmania- 58 Kalyan S, Kabelitz D. When neutrophils meet T cells: beginnings
sis? Br J Dermatol 2008; 158:50–8. of a tumultuous relationship with underappreciated potential.
41 Dabiri S, Hayes MM, Meymandi SS, Basiri M, Soleimani F, Eur J Immunol 2014; 44:627–33.
Mousavi MR. Cytologic features of ‘dry-type’ cutaneous leish- 59 Sabbione F, Gabelloni ML, Ernst G et al. Neutrophils suppress
maniasis. Diagn Cytopathol 1998; 19:182–5. gammadelta T-cell function. Eur J Immunol 2014; 44:819–30.
42 Daboul MW. Role of neutrophils in cutaneous leishmaniasis. 60 Afonso L, Borges VM, Cruz H et al. Interactions with apoptotic
East Mediterr Health J 2010; 16:1055–8. but not with necrotic neutrophils increase parasite burden in
43 Dantas ML, Oliveira JM, Carvalho L et al. Comparative analysis of human macrophages infected with Leishmania amazonensis.
the tissue inflammatory response in human cutaneous and disse- J Leukoc Biol 2008; 84:389–96.
minated leishmaniasis. Mem Inst Oswaldo Cruz 2014; 109:202–9. 61 Ribeiro-Gomes FL, Otero AC, Gomes NA et al. Macrophage
44 Boaventura VS, Santos CS, Cardoso CR et al. Human mucosal interactions with neutrophils regulate Leishmania major infec-
leishmaniasis: neutrophils infiltrate areas of tissue damage that tion. J Immunol 2004; 172:4454–62.
express high levels of Th17-related cytokines. Eur J Immunol 62 de Souza Carmo EV, Katz S, Barbieri CL. Neutrophils reduce
2010; 40:2830–6. the parasite burden in Leishmania (Leishmania) amazonensis-
45 Beil WJ, Meinardus-Hager G, Neugebauer DC, Sorg C. Differen- infected macrophages. PLoS One 2010; 5:e13815.
ces in the onset of the inflammatory response to cutaneous 63 Novais FO, Santiago RC, Bafica A et al. Neutrophils and
leishmaniasis in resistant and susceptible mice. J Leukoc Biol macrophages cooperate in host resistance against Leishmania
1992; 52:135–42. braziliensis infection. J Immunol 2009; 183:8088–98.
46 Muller K, van Zandbergen G, Hansen B et al. Chemokines, nat- 64 Carlsen ED, Jie Z, Liang Y et al. Interactions between neutrophils
ural killer cells and granulocytes in the early course of Leishma- and Leishmania braziliensis amastigotes facilitate cell activation
nia major infection in mice. Med Microbiol Immunol 2001; and parasite clearance. J Innate Immun 2015; 7:354–63.
190:73–6. 65 Mougneau E, Bihl F, Glaichenhaus N. Cell biology and immu-
47 Thalhofer CJ, Chen Y, Sudan B, Love-Homan L, Wilson ME.
nology of Leishmania. Immunol Rev 2011; 240:286–96.
Leukocytes infiltrate the skin and draining lymph nodes in
66 Bertrand A, Jourdan J, Prefaut C. Granulopenia of infectious
response to the protozoan Leishmania infantum chagasi. Infect
origin. Sem Hop 1975; 51:667–76.
Immun 2011; 79:108–17.
67 Costa CH, Werneck GL, Costa DL et al. Is severe visceral leish-
48 Carlsen ED, Hay C, Henard CA, Popov V, Garg NJ, Soong L.
maniasis a systemic inflammatory response syndrome? A case
Leishmania amazonensis amastigotes trigger neutrophil activa-
control study. Rev Soc Bras Med Trop 2010; 43:386–92.
tion but resist neutrophil microbicidal mechanisms. Infect
68 Musumeci S, D’Agata A, Schiliro G, Fischer A. Studies of the
Immun 2013; 81:3966–74.
neutropenia in kala-azar: results in two patients. Trans R Soc
49 Guimaraes-Costa AB, Nascimento MT, Froment GS et al. Leish-
Trop Med Hyg 1976; 70:500–3.
mania amazonensis promastigotes induce and are killed by neu-
69 Pollack S, Nagler A, Liberman D et al. Immunological studies
trophil extracellular traps. Proc Natl Acad Sci USA 2009; 106:
of pancytopenia in visceral leishmaniasis. Isr J Med Sci 1988;
6748–53.
24:70–4.
50 Ritter U, Frischknecht F, van Zandbergen G. Are neutrophils
70 Badaro R, Nascimento C, Carvalho JS et al. Recombinant
important host cells for Leishmania parasites? Trends Parasitol
human granulocyte-macrophage colony-stimulating factor
2009; 25:505–10.
reverses neutropenia and reduces secondary infections in vis-
51 van Zandbergen G, Klinger M, Mueller A et al. Cutting edge:
ceral leishmaniasis. J Infect Dis 1994; 170:413–8.
neutrophil granulocyte serves as a vector for Leishmania entry
71 Almeida BF, Narciso LG, Bosco AM et al. Neutrophil dysfunc-
into macrophages. J Immunol 2004; 173:6521–5.
52 Ribeiro-Gomes FL, Roma EH, Carneiro MB, Doria NA, Sacks tion varies with the stage of canine visceral leishmaniosis. Vet
DL, Peters NC. Site-dependent recruitment of inflammatory Parasitol 2013; 196:6–12.
cells determines the effective dose of Leishmania major. Infect 72 Shi C, Hohl TM, Leiner I, Equinda MJ, Fan X, Pamer EG.
Immun 2014; 82:2713–27. Ly6G1 neutrophils are dispensable for defense against systemic
53 Peters NC, Kimblin N, Secundino N, Kamhawi S, Lawyer P, Listeria monocytogenes infection. J Immunol 2011; 187:5293–8.
Sacks DL. Vector transmission of Leishmania abrogates vaccine- 73 Archer NK, Harro JM, Shirtliff ME. Clearance of Staphylococcus
induced protective immunity. PLOS Pathog 2009; 5:e1000484. aureus nasal carriage is T cell dependent and mediated through
54 Schuster S, Hurrell B, Tacchini-Cottier F. Crosstalk between interleukin-17A expression and neutrophil influx. Infect Immun
neutrophils and dendritic cells: a context-dependent process. 2013; 81:2070–5.
J Leukoc Biol 2013; 94:671–5. 74 Ribes S, Regen T, Meister T et al. Resistance of the brain to
55 Silva MT. When two is better than one: macrophages and neu- Escherichia coli K1 infection depends on MyD88 signaling and
trophils work in concert in innate immunity as complementary the contribution of neutrophils and monocytes. Infect Immun
and cooperative partners of a myeloid phagocyte system. 2013; 81:1810–9.
J Leukoc Biol 2010; 87:93–106. 75 Wang JX, Bair AM, King SL et al. Ly6G ligation blocks recruit-
56 Costantini C, Cassatella MA. The defensive alliance between ment of neutrophils via a beta2-integrin-dependent mechanism.
neutrophils and NK cells as a novel arm of innate immunity. Blood 2012; 120:1489–98.
J Leukoc Biol 2011; 89:221–33. 76 Lima GM, Vallochi AL, Silva UR, Bevilacqua EM, Kiffer MM,
57 Jaeger BN, Donadieu J, Cognet C et al. Neutrophil depletion Abrahamsohn IA. The role of polymorphonuclear leukocytes in
impairs natural killer cell maturation, function, and homeosta- the resistance to cutaneous Leishmaniasis. Immunol Lett 1998;
sis. J Exp Med 2012; 209:565–80. 64:145–51.

C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V 117
E. D. Carlsen et al.

77 Smelt SC, Cotterell SE, Engwerda CR, Kaye PM. B cell-deficient during Leishmania braziliensis infection in mice. Infect Immun
mice are highly resistant to Leishmania donovani infection, but 2009; 77:2948–56.
develop neutrophil-mediated tissue pathology. J Immunol 2000; 89 Soong L, Henard CA, Melby PC. Immunopathogenesis of
164:3681–8. non-healing American cutaneous leishmaniasis and progres-
78 Rousseau D, Demartino S, Ferrua B et al. In vivo involvement sive visceral leishmaniasis. Semin Immunopathol 2012; 34:
of polymorphonuclear neutrophils in Leishmania infantum 735–51.
infection. BMC Microbiol 2001; 1:17. 90 Vargas-Inchaustegui DA, Xin L, Soong L. Leishmania braziliensis
79 Sousa LM, Carneiro MB, Resende ME et al. Neutrophils have a infection induces dendritic cell activation, ISG15 transcription,
protective role during early stages of Leishmania amazonensis and the generation of protective immune responses. J Immunol
infection in BALB/c mice. Parasite Immunol 2014; 36:13–31. 2008; 180:7537–45.
80 Hurrell BP, Schuster S, Grun E et al. Rapid sequestration of 91 Tenorio Mda S, Oliveira e Sousa L, Paixao Mdos S et al. Vis-
Leishmania mexicana by neutrophils contributes to the develop- ceral leishmaniasis in a captive crab-eating fox Cerdocyon thous.
ment of chronic lesion. PLOS Pathog 2015; 11:e1004929. J Zoo Wildl Med 2011; 42:608–16.
81 Nakano H, Yanagita M, Gunn MD. CD11c1B2201Gr-11 cells in 92 Liu C, Desikan R, Ying Z et al. Effects of a novel pharmacologic
mouse lymph nodes and spleen display characteristics of plas- inhibitor of myeloperoxidase in a mouse atherosclerosis model.
macytoid dendritic cells. J Exp Med 2001; 194:1171–8. PLOS ONE 2012; 7: e50767.
82 Geissmann F, Jung S, Littman DR. Blood monocytes consist of 93 Hazen SL, Zhang R, Shen Z et al. Formation of nitric oxide-
two principal subsets with distinct migratory properties. Immu- derived oxidants by myeloperoxidase in monocytes: pathways
nity 2003; 19:71–82. for monocyte-mediated protein nitration and lipid peroxidation
83 Tepper RI, Coffman RL, Leder P. An eosinophil-dependent in vivo. Circ Res 1999; 85:950–8.
mechanism for the antitumor effect of interleukin-4. Science 94 Rosas M, Thomas B, Stacey M, Gordon S, Taylor PR. The mye-
1992; 257:548–51. loid 7/4-antigen defines recently generated inflammatory macro-
84 Matsuzaki J, Tsuji T, Chamoto K, Takeshima T, Sendo F, phages and is synonymous with Ly-6B. J Leukoc Biol 2010; 88:
Nishimura T. Successful elimination of memory-type CD81 T 169–80.
cell subsets by the administration of anti-Gr-1 monoclonal anti- 95 Sadick MD, Locksley RM, Tubbs C, Raff HV. Murine cutaneous
body in vivo. Cell Immunol 2003; 224:98–105. leishmaniasis: resistance correlates with the capacity to generate
85 Tasew G, Nylen S, Lieke T et al. Systemic FasL and TRAIL neu- interferon-gamma in response to Leishmania antigens in vitro.
tralisation reduce leishmaniasis induced skin ulceration. PLOS J Immunol 1986; 136:655–61.
Negl Trop Dis 2010; 4:e844. 96 Locksley RM, Heinzel FP, Sadick MD, Holaday BJ, Gardner KD
86 Souza VL, Veras PS, Welby-Borges M et al. Immune and inflam- Jr. Murine cutaneous leishmaniasis: susceptibility correlates with
matory responses to Leishmania amazonensis isolated from dif- differential expansion of helper T-cell subsets. Ann Inst Pasteur
ferent clinical forms of human leishmaniasis in CBA mice. Mem Immunol 1987; 138:744–9.
Inst Oswaldo Cruz 2011; 106:23–31. 97 Soong L, Chang CH, Sun J et al. Role of CD41 T cells in
87 Guerra JA, Prestes SR, Silveira H et al. Mucosal Leishmaniasis pathogenesis associated with Leishmania amazonensis infection.
caused by Leishmania (Viannia) braziliensis and Leishmania J Immunol 1997; 158:5374–83.
(Viannia) guyanensis in the Brazilian Amazon. PLOS Negl Trop 98 Qi H, Ji J, Wanasen N, Soong L. Enhanced replication of Leish-
Dis 2011; 5:e980. mania amazonensis amastigotes in gamma interferon-stimulated
88 Vargas-Inchaustegui DA, Tai W, Xin L, Hogg AE, Corry DB, murine macrophages: implications for the pathogenesis of cuta-
Soong L. Distinct roles for MyD88 and Toll-like receptor 2 neous leishmaniasis. Infect Immun 2004; 72:988–95.

118 C 2015 British Society for Immunology, Clinical and Experimental Immunology, 182: 109–118
V

You might also like