ArticlePDF Available

Vaccination with L. infantum chagasi Nucleosomal Histones Confers Protection against New World Cutaneous Leishmaniasis Caused by Leishmania braziliensis

PLOS
PLOS ONE
Authors:

Abstract and Figures

Nucleosomal histones are intracellular proteins that are highly conserved among Leishmania species. After parasite destruction or spontaneous lysis, exposure to these proteins elicits a strong host immune response. In the present study, we analyzed the protective capability of Leishmania infantum chagasi nucleosomal histones against L. braziliensis infection using different immunization strategies. BALB/c mice were immunized with either a plasmid DNA cocktail (DNA) containing four Leishmania nucleosomal histones or with the DNA cocktail followed by the corresponding recombinant proteins plus CpG (DNA/Protein). Mice were later challenged with L. braziliensis, in the presence of sand fly saliva. Lesion development, parasite load and the cellular immune response were analyzed five weeks after challenge. Immunization with either DNA alone or with DNA/Protein was able to inhibit lesion development. This finding was highlighted by the absence of infected macrophages in tissue sections. Further, parasite load at the infection site and in the draining lymph nodes was also significantly lower in vaccinated animals. This outcome was associated with increased expression of IFN-γ and down regulation of IL-4 at the infection site. The data presented here demonstrate the potential use of L. infantum chagasi nucleosomal histones as targets for the development of vaccines against infection with L. braziliensis, as shown by the significant inhibition of disease development following a live challenge.
Content may be subject to copyright.
Vaccination with
L. infantum chagasi
Nucleosomal
Histones Confers Protection against New World
Cutaneous Leishmaniasis Caused by
Leishmania
braziliensis
Marcia W. Carneiro
1
, Diego M. Santos
1
, Kiyoshi F. Fukutani
1
, Jorge Clarencio
1
, Jose Carlos Miranda
1
,
Claudia Brodskyn
1,2
, Aldina Barral
1,2
, Manoel Barral-Netto
1,2
, Manuel Soto
3
, Camila I. de Oliveira
1,2
*
1 Centro de Pesquisas Gonc¸alo Moniz, The Oswaldo Cruz Foundation, Salvador, Brazil, 2 Instituto de Investigac¸a
˜
o em Imunologia, Salvador, Brazil, 3 Centro de Biologı
´
a
Molecular Severo Ochoa, Consejo Superior de Investigaciones Cientificas, Departamento de Biologia Molecular, Universidad Autonoma de Madrid, Madrid, Spain
Abstract
Background:
Nucleosomal histones are intracellular proteins that are highly conserved among Leishmania species. After
parasite destruction or spontaneous lysis, exposure to these proteins elicits a strong host immune response. In the present
study, we analyzed the protective capability of Leishmania infantum chagasi nucleosomal histones against L. braziliensis
infection using different immunization strategies.
Methodology/Principal Findings:
BALB/c mice were immunized with either a plasmid DNA cocktail (DNA) containing four
Leishmania nucleosomal histones or with the DNA cocktail followed by the corresponding recombinant proteins plus CpG
(DNA/Protein). Mice were later challenged with L. braziliensis, in the presence of sand fly saliva. Lesion development, parasite
load and the cellular immune response were analyzed five weeks after challenge. Immunization with either DNA alone or
with DNA/Protein was able to inhibit lesion development. This finding was highlighted by the absence of infected
macrophages in tissue sections. Further, parasite load at the infection site and in the draining lymph nodes was also
significantly lower in vaccinated animals. This outcome was associated with increased expression of IFN-c and down
regulation of IL-4 at the infection site.
Conclusion:
The data presented here demonstrate the potential use of L. infantum chagasi nucleosomal histones as targets
for the development of vaccines against infection with L. braziliensis, as shown by the significant inhibition of disease
development following a live challenge.
Citation: Carneiro MW, Santos DM, Fukutani KF, Clarencio J, Miranda JC, et al. (2012) Vaccination with L. infantum chagasi Nucleosomal Histones Confers
Protection against New World Cutaneous Leishmaniasis Caused by Leishmania braziliensis. PLoS ONE 7(12): e52296. doi:10.1371/journal.pone.0052296
Editor: Dario S. Zamboni, University of Sa
˜
o Paulo, Brazil
Received August 14, 2012; Accepted November 12, 2012; Published December 20, 2012
Copyright: ß 2012 Carneiro et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by grants from Conselho Nacional de Desenvolvimento Cientı
´
fico e Tecnolo
´
gico and CYTED. The funders had no role in study
design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: camila@bahia.fiocruz.br
Introduction
Leishmaniasis is an infectious disease with significant economic
impact in several countries. Over three hundred million people are
exposed to the parasites, with 12 million infected worldwide,
predominantly in tropical and subtropical countries (World Health
Organization page: http://www.who.int/emc/diseases/leish/
leisdis1.html). Leishmaniasis can be caused by different species
of Leishmania spp. protozoans that infect macrophages in the human
host. The treatments available for all forms of leishmaniasis are
toxic, and drug resistance is on the rise, further increasing the need
for vaccine development [1].
Numerous attempts have been made to find a protective antigen
against leishmaniasis and several candidates have been tested for
this purpose [2,3], including histones. Histones are structural
proteins found in the nucleus, where they play an important role in
the organization and function of chromatin. There are five main
classes of histones; four of them (H2A, H2B, H3 and H4) form the
nucleosomal core unit of chromatin, whereas H1 joins to linker
DNA. The percentage of similarity between Leishmania nucleosome
forming histones and their mammal counterparts ranges from
49% (for the H2B) to 63% (for the H3) [4]. Differences are mainly
located in the aminoacid sequences of the nucleosome-exposed
tails of the four histones [5]. So far, no cross reactivity was found
between Leishmania histones and their mammalian counterparts.
Antibodies specific for parasite histones, obtained from dogs with
visceral leishmaniasis, react against Leishmania H2A [6], H3 [7],
H2B and H4 [8] but do not recognize mammalian histones.
Antibodies in sera from patients with cutaneous or mucocutaneous
leishmaniasisis also recognize parasite histone H1 but not the
human counterpart [9]. Regarding the T cell immunogenicity of
parasite histones, recombinant versions of H2B [10] or H2A [11]
induced IFN-c secretion upon stimulation of PBMCs obtained
from cutaneous leishmaniasis patients. The T cell response was
PLOS ONE | www.plosone.org 1 December 2012 | Volume 7 | Issue 12 | e52296
specific for parasite histones, since T cell lines derived from
cutaneous leishmaniasis patients did not respond to mammalian
histones [12].
Leishmania histones are recognized by sera from cutaneous
leishmaniasis patients [13] and from dogs infected with Leishmania
[6,7,8] and it has been hypothesized that these parasite proteins
may trigger an immune response after active destruction or
spontaneous cytolysis of Leishmania amastigotes [4,14]. Immuniza-
tion with the histone H1 was able to confer protection against L.
major [15,16] and L. infantum [17]. Immunization with H2B was
able to confer protection in mice infected with L. major [18], as
seen by a decrease in parasite load and lesion size. The protective
effect against leishmaniasis was also observed upon immunization
with the four core histone proteins (H2A, H2B, H3 and H4)
[19,20,21].
In addition to antigen selection, the immunization strategy is
also important for generating protection. Various antigens have
been tested and evaluated as DNA and/or recombinant protein
vaccine candidates in murine models of leishmaniasis, resulting in
various degrees of protection [22]. DNA vaccination has also been
tested in heterologous prime-boost vaccination regimes [23], in
which the immune system is primed with DNA and boosted with
a different formulation of the corresponding antigen. This strategy
has been shown to be effective in experimental models of
cutaneous leishmaniasis [24,25,26].
Based on the protective capacity of the four nucleosomal
histones against L. infantum chagasi infection, we hypothesized that
this antigenic cocktail would confer protection against the
development of New World cutaneous leishmaniasis caused by
L. braziliensis, a species for which there are few published studies
regarding vaccine development in comparison with L. major [27].
The present study also compared immunization strategies in-
volving plasmid DNA only and plasmid DNA plus recombinant
proteins.
Materials and Methods
Mice and Ethics Statement
Female BALB/c mice, 6–8 weeks of age, were obtained from
CPqGM/FIOCRUZ animal facility where they were maintained
under pathogen-free conditions. Animals were randomly distrib-
uted into groups of five and each group was subjected to a specific
immunization strategy. All animal work was conducted according
to the Guidelines for Animal Experimentation of the Cole´gio
Brasileiro de Experimentac¸a˜o Animal and of the Conselho
Nacional de Controle de Experimentac¸a˜o Animal. The local
Ethics Committee on Animal Care and Utilization (CEUA)
approved all procedures involving animals (CEUA Centro de
Pesquisas Gonc¸alo Muniz/FIOCRUZ - L031/08).
Preparation of DNA Plasmids and Recombinant Proteins
The recombinant plasmids [21](pcDNA3-LiH2A, pcDNA3-
LiH2B, pcDNA3-LiH3 and pcDNA3-LiH4) were prepared
using the endotoxin-free Giga-preparation Kit (Qiagen) follow-
ing the manufacturer’s instructions. The final pellet was
resuspended in sterile water and stored at 220uC until use.
Expression and purification of the His-tagged recombinant
proteins (pQEH2A, pQE-H2B, pQE-H3 and pQE-H4) were
performed as previously described [21]. After binding to a Ni-
NTA agarose column (Qiagen), recombinant proteins were
gradually refolded on the affinity column as described [28].
Recombinant proteins were eluted with 0.3 M imidazole and
dialyzed against PBS. Finally, proteins were passed through
a polymyxin-agarose column (Sigma) in order to eliminate
endotoxins. Residual endotoxin was measured with Quantitative
Chromogenic Limulus Amebocyte assay (QCL-1000, BioWhit-
taker), showing that recombinant histone preparations were
essentially endotoxin-free (less than 30 ng endotoxin per mg of
recombinant protein).
Immunization Strategies
BALB/c mice were immunized three times, with a two-week
interval between each immunization. Animals received three
intramuscular (i.m.) injections of a DNA (100 mg) cocktail
containing 25 mg of each recombinant plasmid (pcDNA3-LiH2A,
pcDNA3-LiH2B, pcDNA3-LiH3 and pcDNA3-LiH4) in a total
volume of 50 ml PBS. Control mice were immunized (i.m.) with
100 mg of WT pcDNA3. Alternatively, mice received two
inoculations (i.m.) of the recombinant DNA cocktail followed by
a third (s.c.) inoculation of a recombinant protein (20 mg) cocktail
containing 5 mg of each recombinant protein (H2A, H2B, H3 and
H4) and 25 mg of each CpG ODN (59- tcagcgttga-39 and 59-
gctagcgttagcgt-39) (E-OLIGOS). Control mice received two
immunizations (i.m.) with WT pcDNA3 followed by a third
inoculation (s.c.) of saline+CpG. DNA immunizations were
performed in the left quadriceps and protein immunizations were
performed in the left footpad.
Cytokine Detection in Immunized Mice
BALB/c mice were immunized as described above. Two weeks
after the last immunization, mice were euthanized and single-cell
suspensions of draining lymph nodes (dLN) were prepared
aseptically. Briefly, dLN were homogenized in RPMI 1640 and
cells were resuspended in RPMI supplemented with 2 mM L-
glutamine, 100 U/mL penicillin, 100 mg/mL streptomycin, 10%
FCS (all from Invitrogen) and 0.05 M b-mercaptoethanol. Cell
suspensions were stimulated for 48 h with 5 mg/mL Concanavalin
A (Amersham Biosciences) or 3 mg/mL of each of the following
recombinant proteins: LiH2A, LiH2B, LiH3 and LiH4. Culture
supernatants were harvested and cytokine concentrations were
assayed using a Th1/Th2 cytokine Cytometric Bead Array (BD
Biosciences), which detects murine IFN-c, IL-4 and TNF-
a according to the manufacturer’s instructions. The data were
acquired and analyzed using a FACSort flow Cytometer (BD
Immunocytometry) and CBA Analysis Software (Becton-Dick-
inson).
Sand Flies and Preparation of SGS
Lutzomyia intermedia salivary glands were obtained as previously
described [29]. Salivary gland from adult female flies were
dissected and transferred to 10 or 20 ml Hepes, 10 mM
pH 7.0 NaCl 0.15 in 1.5 polypropilene vials, usually in groups
of 20 pairs of glands in 20 ml of Hepes saline. Salivary glands were
kept at 275uC until needed, when they were disrupted by
sonication using a Branson Sonifier 450 homogenizer (Branson,
Danbury, CT). Salivary gland sonicate (SGS) was centrifuged at
10,0006g for 4 min and the supernatants were used in the
experiments. The level of lipopolysaccharide (LPS) contamination
of SGS preparations was determined using a commercially
available LAL chromogenic kit (QCL-1000; Lonza Biologics).
LPS concentration was ,0.1 ng/ml.
Parasite Culture, Intradermal Inoculation and Lesion
Measurement
L. braziliensis promastigotes (strain MHOM/BR/01/BA788)
[30] were grown in Schneider medium (Sigma), supplemented
with 100 U/ml of penicillin, 100 mg/ml of streptomycin and 10%
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 2 December 2012 | Volume 7 | Issue 12 | e52296
heat-inactivated fetal calf serum (Invitrogen). Two weeks after the
last immunization, mice were inoculated with L. braziliensis,as
described previously [30]. Challenges consisted of inoculation of
stationary-phase promastigotes (10
5
parasites)+SGS (1 mg, equiv-
alent to 1 pair of salivary glands), in 10 ml of saline. Lesion size was
monitored weekly for 10 weeks, using a digital caliper (Thomas
Scientific).
Parasite Load Estimate
Parasite load was determined using a quantitative limiting
dilution assay as described previously [31]. Animals were
euthanized five weeks post infection. Infected ears and draining
lymph nodes (dLNs) were aseptically excised and homogenized in
Schneider medium (Sigma). The homogenates were serially
diluted in Schneider medium supplemented with 100 U/ml of
penicillin, 100 mg/ml of streptomycin, 10% heat-inactivated fetal
calf serum (all from Invitrogen) and seeded into 96-well plates
containing biphasic blood agar (Novy-Nicolle-McNeal) medium.
The number of viable parasites was determined from the highest
to lowest dilution at which promastigotes could be grown after one
week of incubation at 25uC.
Histology
BALB/c mice were immunized and challenged as described
above. Five weeks post challenge, animals were euthanized and
ears were removed and fixed in 10% formaldehyde. Following
fixation, tissues were processed, embedded in paraffin and 5 mm
sections were stained with hematoxylin and eosin (H & E) and
analyzed by light microscopy.
RNA Isolation and Real-time PCR
Five weeks following infection with L. braziliensis+SGS, mice
were euthanized and infected ears were excised and mechanically
lysed with ceramic beads in a MagNALyzerH instrument (Roche
Molecular Systems) according to manufacturer’s instructions.
Total RNA was extracted from the resulting tissue lysates using
the RNeasy Protect Mini Kit (Qiagen) according to the
manufacturer’s instructions. RNA was eluted in 20 ml water and
used for cDNA synthesis. Real-time PCR was performed on the
ABI Prism 7500 (Applied Biosystems). Thermal cycle conditions
consisted of a two-minute initial incubation at 50uC followed by
a 10 minute denaturation at 95uC and 50 cycles at 95uC for 15
seconds and 60uC for one minute each. Each sample and the
negative control were analyzed in triplicate for each run. The
comparative method was used to analyze gene expression.
Cytokine cycle threshold (C
t
) values were normalized to GAPDH
expression as determined by the equation DC
t
=C
t (cytokine)
–C
t
(GAPDH)
. Fold change was determined by 2
DDCt
, where
DDC
t
= DC
t (experimental)
DC
t (control).
[32]. Primers employed
herein are described elsewhere [33].
Intracellular Cytokine Detection by Flow Cytometry
Reagents for staining cell surface markers and intracellular
cytokines were purchased from BD Biosciences. Measurement of
in vitro cytokine production was performed as described [30].
Briefly, animals were euthanized five weeks post infection. dLNs
were aseptically excised and homogenized in RPMI supple-
mented with 100 U/ml of penicillin, 100 mg/ml of streptomy-
cin, 10% heat-inactivated fetal calf serum (all from Invitrogen).
Cells were then activated using 5 mg/ml Concanavalin A
(Amersham Biosciences) and incubated with 10 mg/ml Brefeldin
A (Sigma). Cells were blocked with anti-Fc receptor antibody
(2.4G2) and were double stained with anti-mouse surface CD4
(L3T4) and CD8 (53–6.7) conjugated to FITC and Cy-Chrome,
respectively. For intracellular staining of cytokines, cells were
permeabilized using Cytofix/Cytoperm and were incubated with
the following anti-cytokine antibodies conjugated to PE: IFN-c
(XMG1.2), IL-4 (BVD4-1D11) or IL-10 (JES5-16E3). The
isotype controls used were rat IgG2b (A95-1) and rat IgG2a
(R35–95). Data were collected and analyzed using CELLQuest
software and a FACSort flow cytometer (Becton Dickinson
Immunocytometry System).
Statistical Analysis
The data are presented as the mean 6 SEM. Comparisons
between four groups (DNA, DNA/Protein and controls) were
performed by Kruskal-Wallis (non-parametric test) followed by
Dunn’s multiple comparisons test. Comparisons between two
groups (DNA vs. control or DNA/Protein vs. control) were
performed by Mann-Whitney (non-parametric t-test) using
GraphPad version 6.0a (Prism) and P-values ,0.05 were
considered significant. To evaluate disease burden in mice, ear
thickness of mice following challenge was recorded weekly for each
individual mouse. The course of disease for experimental and
control mice was plotted individually. The Area under the curve
(AUC) obtained for each mouse immunized with antigen versus
AUC obtained for each control mouse was analyzed by Mann-
Whitney (non-parametric t-test).
Results
Immunization with Nucleosomal Histones Prevents
Lesion Development in Mice Infected by L. braziliensis
Plus Sand Fly Saliva
We first analyzed the immune response induced upon
immunization with a plasmid DNA cocktail encoding histones
H2A, H2B, H3 and H4. Mice inoculated with the recombinant
DNA cocktail (rDNA) did not show a significant increase in IFN-c
(Fig. 1A), IL-4 (Fig. 1B) or TNF-a (Fig. 1C) when compared with
mice immunized with WT DNA. Similar results were observed
regarding antigen specific cytokine production in mice immunized
with the combination of plasmid DNA followed by recombinant
proteins (rDNA/rProtein+CpG) vs. control (WT DNA/CpG)
(Fig. 1D–F). Two weeks after the last immunization, mice were
challenged in the dermis of the ear by co-inoculation of L.
braziliensis plus sand fly (L. intermedia) saliva, in order to mimic the
natural context of infection by Leishmania parasites. Following
challenge, lesion development was monitored for ten weeks. Mice
immunized with rDNA did not develop disease, as shown by
maintenance of ear thickness close to baseline levels (Fig. 2A),
indicating disease prevention. Control mice (immunized with WT
DNA) developed lesions that peaked at five weeks after infection
(Fig. 2A), characteristic of the inoculation of L. braziliensis into the
ear dermis of BALB/c mice [30]. At this time point, ear thickness
reached a maximum of 1.2 mm. Mice inoculated with rDNA/
rProtein+CpG (Fig. 2B) also did not develop lesions whereas
controls immunized with WT DNA/CpG behaved as mice
inoculated with WT DNA (Fig. 2A). Importantly, disease de-
velopment, as determined by the area under the curves (AUCs)
shown in Fig. 2A and Fig. 2B (see Materials and Methods), was
significantly inhibited in mice immunized with rDNA or with
rDNA/rProtein+CpG, when compared with controls (Fig. 2C).
The AUCs of immunized mice (either rDNA only or rDNA/
rProtein+CpG) and of control mice (WT DNA only or WT DNA/
CpG) were similar. This suggests that immunization with L.
infantum chagasi histones inhibit cutaneous leishmaniasis caused by
L. braziliensis.
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 3 December 2012 | Volume 7 | Issue 12 | e52296
We also analyzed ear sections, obtained five weeks after co-
inoculation with L. braziliensis plus sand fly (L. intermedia) saliva. As
shown in Fig. 3A, mice immunized with WT DNA developed an
intense inflammatory infiltrate with an accumulation of parasite-
infected macrophages. In mice immunized with WT DNA,
infected cells were abundant (Fig. 3A), differently from mice
immunized with rDNA (Fig. 3B). In tissue sections from mice
immunized with WT DNA/CpG, we observed a dense and
widespread inflammatory infiltrate containing infected macro-
phages displaying a foamy aspect (Fig. 3C). In rDNA/rPro-
tein+CpG-inoculated mice, amastigotes were not detected and the
inflammatory infiltrate was characterized by the presence of rare
eosinophils, plasmocytes and epithelioid macrophages (Fig. 3D).
Parasite Load and Cytokine Expression Profile at the
Infection Site
Given the significant inhibition of lesion development following
immunization with plasmid rDNA (Fig. 2A and C) or with rDNA/
rProtein+CpG (Fig. 2B and C), we also investigated the parasite
load. At five weeks post parasite inoculation, when ear thickness
was at its peak (Fig. 2A and B), mice immunized with rDNA
displayed a significantly lower parasite load at the infection site
(Fig. 4A) and at the draining lymph node (Fig. 4B). Comparable
results were observed with rDNA/rProtein+CpG-immunized mice
(Fig. 4A and B), in terms of inhibition in parasite replication. The
parasite load detected in the ear of mice immunized with histones
(rDNA vs. rDNA/rProtein+CpG) or in control animals (WT DNA
vs. WT DNA/CpG) were not significantly different (Fig. 4A and
B).
Figure 1. Antigen specific cytokine production following immunization with nucleosomal histones. BALB/c mice (5 per group) were
immunized with wild type DNA (WT DNA) or with recombinant DNA coding for nucleosomal histones (rDNA) (A–C). Alternatively, BALB/c mice (5 per
group) were immunized with wild type DNA followed by CpG (WT DNA+CpG), or with recombinant DNA followed by recombinant nucleosomal
histones+CpG (rDNA/rProtein+CpG) (D–F). Two weeks after the last immunization, dLNs were collected and cells were re-stimulated with the
recombinant proteins or with Concanavalin A (Con A). Antigen specific cytokine production in culture supernatants was determined by flow
cytometry, using a Th1–Th2 Cytometric Bead Array. Data are presented as the mean+SEM and are from two independent experiments.
doi:10.1371/journal.pone.0052296.g001
Figure 2. Lesion development in mice immunized with nucleosomal histones following infection with
L. braziliensis
plus sand fly
saliva. BALB/c mice (5 per group) were immunized with WT DNA or with rDNA (A). Alternatively, BALB/c mice (5 per group) were immunized with WT
DNA+CpG or with rDNA/rProtein+CpG (B). Two weeks after the last immunization, mice were challenged with L. braziliensis+sand fly saliva. The
course of lesion development was monitored weekly and bars represent the means and standard errors from two independent experiments. The
areas contained underneath the curves obtained in (A) and in (B) for each individual mouse from experimental and control groups were compared
(C). Data are presented as the mean+SEM.
doi:10.1371/journal.pone.0052296.g002
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 4 December 2012 | Volume 7 | Issue 12 | e52296
Figure 3. Histological aspects of ear lesions in mice immunized with nucleosomal histones and challenged with
L. braziliensis
plus
sand fly saliva. BALB/c mice (five mice per group) were immunized with WT DNA (A) or with rDNA (B). Alternatively, mice were immunized with WT
DNA/CpG (C) or with rDNA/rProtein+CpG (D). Two weeks after the last inoculation, mice were challenged with L. braziliensis+sand fly saliva. Ears were
removed at 5 weeks post infection and stained with hematoxylin and eosin. Panels represent 1006magnification. (C) Dark symbols indicate infected
macrophages displaying a foamy aspect. (D) Red symbols indicate plasmocytes and dashed arrows indicate eosinophils.
doi:10.1371/journal.pone.0052296.g003
Figure 4. Parasite load following immunization with nucleosomal histones and challenge with
L. braziliensis
plus sand fly saliva.
BALB/c mice (5 per group) were immunized with WT DNA or with rDNA. Alternatively, BALB/c mice (5 per group) were immunized with rDNA/
rProtein+CpG. Two weeks after the last immunization, mice were challenged with L. braziliensis+sand fly saliva. Ear (A) and draining lymph node (B)
parasite loads were determined five weeks post infection via a limiting dilution assay. Data are presented as the mean+SEM and are from two
independent experiments.
doi:10.1371/journal.pone.0052296.g004
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 5 December 2012 | Volume 7 | Issue 12 | e52296
Therefore, immunization with L. infantum chagasi nucleosomal
histones modified the course of infection in mice challenged with
L. braziliensis.
Based on these results, we analyzed cytokine expression at the
infection site. Total RNA was obtained from ear sections five
weeks after parasite inoculation, when ear thickness was at its peak
in control mice (Fig. 2) and the parasite load was significantly
different between immunized versus control groups (Fig. 4). RNA
was subjected to real-time PCR analysis, and cytokine gene
expression was normalized to GAPDH (housekeeping gene), as
described in the Materials and Methods. Ears from rDNA-
immunized mice showed a two-fold up-regulation in IFN-c
expression in comparison with control mice (Fig. 5A) whereas
the expression of IL-4 and IL-10 were down-regulated (Fig. 5A).
Mice immunized with rDNA/rProtein+CpG also showed up-
regulation of IFN-c expression (Fig. 5B). These results indicate the
predominance of a Th1-polarized response and can be correlated
with the decreased disease burden (Fig. 2) and the lower parasite
load observed at the infection site (Fig. 4) of mice immunized with
nucleosomal histones.
Cellular Immune Response in dLNs
Based on the lower parasite load observed in the dLNs of
immunized mice (Fig. 2), we also evaluated the presence of
cytokine-secreting cells therein. Five weeks after infection, cells
from lymph nodes draining the infection site were re-stimulated
in vitro with recombinant histones and the frequency of cytokine-
secreting cells was determined by flow cytometry (Figure S1). The
percentage of CD4+ IFN-c-secreting cells was similar in control vs.
rDNA-immunized mice (Fig. 6A) and in controls vs. rDNA/
rProtein+CpG-immunized mice (Fig. 6B). Also, we did not detect
significant differences in the percentage of CD4+ IL-4+-secreting
cells in (Fig. 6C–D) or in the frequency of CD4+IL-10+-secreting
cells (Fig. 6E–F), when comparing the two immunization
strategies. The percentage of CD8+ IFN-c-secreting cells was
similar in control vs. rDNA-immunized mice (Fig. 7A) but was
significantly higher in mice immunized with rDNA/rPro-
tein+CpG when compared with controls (Fig. 7B). The frequency
of CD8+IL-4+-secreting cells was also similar in control vs. rDNA-
immunized mice (Fig. 7C) but was significantly lower in mice
immunized with rDNA/rProtein+CpG when compared with
controls (Fig. 7D). As seen with CD4+ cells, the frequency of
CD8+IL-10+-secreting cells did not differ significantly in control
vs. immunized mice (Fig. 7E and F).
Discussion
In the present work, we evaluated the comparative vaccine
potential of rDNA and rDNA/rProtein+CpG using L. infantum
chagasi histones H2A, H2B, H3 and H4 in the context of eliciting
immunity against L. braziliensis. The results reported here suggest
that both strategies were able to prevent lesion development in an
experimental model of New World cutaneous leishmaniasis.
Previous studies using L. infantum chagasi nucleosomal histones
have shown the induction of a Th1-biased response, with
significant levels of protection against L. major infection in mice
[19,20,21]. To evaluate efficacy in an experimental model of New
World Cutaneous Leishmaniasis, we employed two immunization
strategies: rDNA and rDNA/rProtein+CpG. The prime-boost
strategy (rDNA/rProtein+CpG) aims at augmenting immune
responses induced by rDNA vaccination alone and it has been
successfully employed previously in leishmaniasis [34,35,36,37].
Here, recombinant proteins were formulated with CpG, given its
ability to stimulate macrophages and dendritic cells (DCs) to
synthesize cytokines, up-regulate the expression of co-stimulatory
molecules and to enhance the cross-presentation properties of DCs
[38,39]. In the present study, immunization with rDNA alone or
with rDNA followed by rProtein+CpG did not elicit a strong
immune response, as shown by the lack of a significant increase in
cytokine production in immunized mice. Herein, cell cultures were
simultaneously stimulated with the recombinant proteins corre-
sponding to histones H2A, H2B, H3 and H4 whereas in the study
by Iborra et al., cells from mice immunized with nucleosomal
histones were stimulated with each recombinant protein separately
and the authors were able to detect a significant increase in IFN-c
secretion [21].
Despite the fact that we did not detect a robust pre-challenge
immune response, both immunization strategies were able to
significantly inhibit lesion development upon intradermal in-
oculation of L. braziliensis, in the presence of L. intermedia sand fly
saliva. Although mice were challenged with a high inoculum (10
5
stationary-phase promastigote forms), significant protection was
achieved as shown by lack of lesion development and by the
significant reduction (over 2 log) in parasite load observed at the
infection site in mice vaccinated with either rDNA or rDNA/
rProtein+CpG. This level of protection is comparable to
vaccination studies published employing nucleosomal histones
and L. major infection [40]. Regarding sand fly saliva, it contains
pharmacologically active molecules that promote adequate blood
feeding and that may contribute to establishment of infection by
Leishmania (rev. in [41]). Among the salivary components
characterized to date are maxadilan [42], that modulates the
inflammatory response by inhibiting cytokines such as TNF-a;
hyaluronidase, that helps the diffusion of other pharmacological
substances through the skin matrix [43] and a 5-nucleotidase, that
exerts a vasodilator and anti-platelet aggregation role by
converting AMP to adenosine [44]. In the case of L. braziliensis,
co-inoculation of salivary gland sonicate (SGS) and parasites led to
Figure 5. Cytokine expression in the ear dermis following
immunization with nucleosomal histones and challenge with
L.
braziliensis
plus sand fly saliva. BALB/c mice (5 per group) were
immunized with rDNA (A) or with rDNA/rProtein+CpG (B). Two weeks
after the last immunization, mice were challenged with L. brazilien-
sis+sand fly saliva. Relative quantification of IFN-c, IL-4 and IL-10 at the
infection site was carried out five weeks after infection. Cytokine cycle
threshold (C
t
) values were normalized to GAPDH expression (house-
keeping) as determined by DC
t
=C
t (cytokine)
–C
t (GAPDH)
. Fold change
was determined by real-time PCR, using the 2
DDCt
method, where
DDC
t
= DC
t (experimental)
DC
t (control)
(see Materials and Methods). Data
(mean+SEM) are presented as fold increase in gene expression of
immunized mice over control mice and are from two independent
experiments.
doi:10.1371/journal.pone.0052296.g005
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 6 December 2012 | Volume 7 | Issue 12 | e52296
a significant exacerbation of both lesion size and parasite load
[45,46,47]. We later showed that immunization with L. intermedia
SGS altered the course of experimental infection with L. braziliensis
[29] and stimulation of immune mice with a combination of SGS
and L. braziliensis led to a decreased CXCL10 expression and
increased IL-10 expression [33]. These results suggest that L.
intermedia saliva exerts important immunomodulatory activities
and, therefore, we judged important to include salivary compo-
nents to the challenge inoculum. Other studies also employed
salivary gland sonicate to ‘‘mimic’’ the effects of sand fly saliva in
the context of vaccination [48,49,50,51,52]. More recently, Peters
et al. showed that vaccination with autoclaved L. major anti-
gen+CpG confers protection against a needle inoculation of
Figure 6. Intracellular cytokine production by CD4+ cells from
mice immunized with nucleosomal histones and challenged
with
L. braziliensis
plus sand fly saliva. BALB/c mice (5 per group)
were immunized as described. Two weeks after the last immunization,
mice were challenged with L. braziliensis+sand fly saliva. Five weeks
later, draining lymph nodes were pooled and cells were preincubated
with Brefeldin A for four hours before staining. Data represent the
frequency of CD4+ cells positive for IFN-c (A, B), IL-4 (C, D) and IL-10 (D,
E) with signals for the particular cytokine that were greater than the
background signals established using isotype controls. Data are
presented as the mean+SEM and are from two independent experi-
ments.
doi:10.1371/journal.pone.0052296.g006
Figure 7. Intracellular cytokine production by CD8+ cells from
mice immunized with nucleosomal histones and challenged
with
L. braziliensis
plus sand fly saliva. BALB/c mice (5 per group)
were immunized as described. Two weeks after the last immunization,
mice were challenged with L. braziliensis+sand fly saliva. Five weeks
later, draining lymph nodes were pooled and cells were preincubated
with Brefeldin A for four hours before staining. Data represent the
frequency of CD8+ cells positive for IFN-c (A, B), IL-4 (C, D) and IL-10 (D,
E) with signals for the particular cytokine that were greater than the
background signals established using isotype controls. Data are
presented as the mean+SEM and are from two independent experi-
ments.
doi:10.1371/journal.pone.0052296.g007
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 7 December 2012 | Volume 7 | Issue 12 | e52296
parasites but not against challenge with infected sand flies [53]
whereas KSAC, a polyprotein vaccine candidate [54], conferred
protection against the bite of L. major-infected sand flies [55]. To
date, a colony of L. intermedia sand flies is not available, hampering
the possibility of using infected sand flies in challenge experiments.
In this sense, we believe that addition of SGS at the time of
challenge partially addresses this limitation and allows us to
evaluate the effects of salivary molecules in the context of
vaccination.
Inhibition of lesion development in mice immunized with
nucleosomal histones (rDNA or rDNA/rProtein+CpG) was
accompanied by a significant decrease in parasite load at the
infection site. These results may be related to a significant up-
regulation of the expression of IFN-c, as seen in infected ears five
weeks after infection. Macrophages control Leishmania infection by
inducing reactive oxygen species when activated by IFN-c [56].
Therefore, we can speculate that IFN-c-secreting cells may have
migrated to the infection site, promoting macrophage activation
and parasite killing. Additionally, Iborra et al. showed 1.22% of
CD4+ IFN-c+ and 1.02% of CD8+ IFN-c+ T cells in mice
challenged with L. major [21] whereas we detected 1.7%
CD4+IFN-c+ (Figure 6) and 1.9% CD8+ IFN-c+ cells (Figure 7),
in mice immunized with rDNA and challenged with L. braziliensis.
We believe these results are comparable in terms of the immune
response detected after challenge and indicate that the protective
capacity of nucleosomal histones is associated with an expansion of
IFN-c-expressing cells.
Chenik et al. documented the participation of the carboxy-
terminal portion of the H2B histone in the activation of Treg cells
[18]. The presence IL-10 secreting cells, with a regulatory
phenotype, may have modulated potentially harmful effects
associated with the development of a Th1-immune response
[57], leading to the control of pathology at the infection site of
mice immunized with nucleossomal histones. Indeed, the presence
of Tregs has been associated with healing of experimental infection
with L. braziliensis [58]. On the other hand, control mice that did
not receive nucleosomal histomes did not develop a Th1-immune
response. In this case, the potential presence of a regulatory
response could have contributed with parasite replication instead
of a control in pathology.
During the past several decades, extensive efforts have been
made to develop an effective Leishmania vaccine [3,22]. The
majority of studies have been conducted with L. major [1]. L.
braziliensis, which is distinguished from other leishmaniasis by its
chronicity, latency and tendency to metastasize in the human host
[59], has been largely neglected in the context of vaccine
development. Moreover, candidate antigens such as the receptor
for activated C kinase protein (LACK), thiol-specific antioxidant
(TSA), Leishmania elongation and initiation factor (LeIF) and L.
major stress-inducible protein 1 (LmST1), all of which induced
protection against L. major, failed to prevent L. braziliensis infection
[60]. These findings highlight the need for continued investigation
into molecules able to confer protection against this particular
species. In conclusion, we believe our results represent an
important contribution to understanding leishmaniasis as they
extend the cross-protective effect of nucleosomal histones from L.
infantum chagasi to a model of New World cutaneous leishmaniasis
caused by L. braziliensis.
Supporting Information
Figure S1 Cytokine expression in CD4+ and in CD8+
cells in mice immunized with nucleossomal histones,
following challenge with L. braziliensis plus sand fly
saliva. BALB/c mice were immunized with DNA coding for
nucleosomal histones and two weeks after the last immunization,
mice were infected in the dermis of the ear with 10
5
L. braziliensis+
sand fly saliva, as described in Materials and Methods. Gates depict
CD4+and CD8+ T lymphocytes present in the draining lymph
node (dLN). The presence of IFN-c
+
, IL-4
+
and IL-10
+
T cells was
determined by flow cytometry in the gated populations. Data
shown are representative dot plots for IFN-c
+,
IL-4
+
and IL-10
labeling.
(TIF)
Acknowledgments
We thank Dr Almerio Noronha for help with histopathology analysis and
documentation. MWC, DMS and KFF were supported by CAPES
fellowships. MBN, AB, CB and CIO are senior investigators from CNPq.
Author Contributions
Conceived and designed the experiments: MWC DMS MS CIO.
Performed the experiments: MWC DMS JC KF JCM. Analyzed the data:
MWC DMS KF JC CB AB MBN MS CIO. Contributed reagents/
materials/analysis tools: MS AB JCM MS. Wrote the paper: MWC MS
CIO.
References
1. Costa CH, Peters NC, Maruyama SR, de Brito EC, Santos IK (2011) Vaccines
for the Leishmaniases: Proposals for a Research Agenda. PLoS Negl Trop Dis 5:
e943.
2. de Oliveira CI, Nascimento IP, Barral A, Soto M, Barral-Netto M (2009)
Challenges and perspectives in vaccination against leishmaniasis. Parasitol Int
58: 319–324.
3. Okwor I, Uzonna J (2009) Vaccines and vaccination strategies against human
cutaneous leishmaniasis. Hum Vaccin 5: 291–230.
4. Requena JM, Alonso C, Soto M (2000) Evolutionarily conserved proteins as
prominent immunogens during Leishmania infections. Parasitol Today 16: 246–
250.
5. Galanti N, Galindo M, Sabaj V, Espinoza I, Toro GC (1998) Histone genes in
trypanosomatids. Parasitol Today 14: 64–70.
6. Soto M, Requena JM, Quijada L, Garcia M, Guzman F, et al. (1995) Mapping
of the linear antigenic determinants from the Leishmania infantum histone H2A
recognized by sera from dogs with leishmaniasis. Immunol Lett 48: 209–214.
7. Soto M, Requena JM, Quijada L, Gomez LC, Guzman F, et al. (1996)
Characterization of the antigenic determinants of the Leishmania infantum
histone H3 recognized by antibodies elicited during canine visceral leishman-
iasis. Clin Exp Immunol 106: 454–461.
8. Soto M, Requena JM, Quijada L, Perez MJ, Nieto CG, et al. (1999) Antigenicity
of the Leishmania infantum histones H2B and H4 during canine viscerocuta-
neous leishmaniasis. Clin Exp Immunol 115: 342–349.
9. Carmelo E, Zurita AI, Martinez E, Valladares B (2006) The sera from
individuals suffering from cutaneous leishmaniasis due to Leishmania brazilensis
present antibodies against parasitic conserved proteins, but not their human
counterparts. Parasite 13: 231–236.
10. Meddeb-Garnaoui A, Toumi A, Ghelis H, Mahjoub M, Louzir H, et al. (2010)
Cellular and humoral responses induced by Leishmania histone H2B and its
divergent and conserved parts in cutaneous and visceral leishmaniasis patients,
respectively. Vaccine 7: 1881–1886.
11. de Carvalho LP, Soto M, Jeronimo S, Dondji B, Bacellar O, et al. (2003)
Characterization of the immune response to Leishmania infantum recombinant
antigens. Microbes Infect 5: 7–12.
12. Probst P, Stromberg E, Ghalib HW, Mozel M, Badaro R, et al. (2001)
Identification and characteriza tion of T cell-stimulating antige ns from
Leishmania by CD4 T cell expression cloning. J Immunol 166: 498–505.
13. Carmelo E, Martinez E, Gonzalez AC, Pinero JE, Patarroyo ME, et al. (2002)
Antigenicity of Leishmania braziliensis histone H1 during cutaneous leishman-
iasis: localization of antigenic determinants. Clin Diagn Lab Immunol 9: 808–
811.
14. Santarem N, Silvestre R, Tavares J, Silva M, Cabral S, et al. (2007) Immune
response regulation by leishmania secreted and nonsecreted antigens. J Biomed
Biotechnol 2007: 85154.
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 8 December 2012 | Volume 7 | Issue 12 | e52296
15. Solioz N, Blum-Tirouvanziam U, Jacquet R, Rafati S, Corradin G, et al. (1999)
The protective capacities of histone H1 against experimental murine cutaneous
leishmaniasis. Vaccine 18: 850–859.
16. Masina S, M MG, Demotz SO, Fasel NJ (2003) Protection against cutaneous
leishmaniasis in outbred vervet monkeys using a recombinant histone H1
antigen. J Infect Dis 188: 1250–1257.
17. Agallou M, Smirlis D, Soteriadou KP, Karagouni E (2012) Vaccination with
Leishmania histone H1-pulsed dendritic cells confers protection in murine
visceral leishmaniasis. Vaccine 30: 5086–5093.
18. Chenik M, Louzir H, Ksontini H, Dilou A, Abdmoule h I, et al. (2006)
Vaccination with the divergent portion of the prote in histone H2B of
Leishmania protects susceptible BALB/c mice against a virulent challenge with
Leishmania major. Vaccine 24: 2521–2529.
19. Carrion J, Folgueira C, Alonso C (2008) Immunization strategies against visceral
leishmaniosis with the nucleosomal histones of Leishmania infantum encoded in
DNA vaccine or pulsed in dendritic cells. Vaccine 26: 2537–2544.
20. Carrion J, Nieto A, Soto M, Alonso C (2007) Adoptive transfer of dendritic cells
pulsed with Leishmania infantum nucleosomal hist ones confers protection
against cutaneous leishmaniosis in BALB/c mice. Microbes Infect 9: 735–743.
21. Iborra S, Soto M, Carrion J, Alonso C, Requena JM (2004) Vaccination with
a plasmid DNA cocktail encoding the nucleosomal histones of Leishmania
confers protection against murine cutaneous leishmaniosis. Vaccine 22: 3865–
3876.
22. Launois P, Tacchini-Cottier F, Kieny M (2008) Cutaneous leishmaniasis:
progress towards a vaccine. Expert Rev Vaccines 7: 1277–1287.
23. Ramshaw IA, Ramsay AJ (2000) The prime-boost strategy: exciting prospects for
improved vaccination. Immunol Today 21: 163–165.
24. Gonzalo RM, del Real G, Rodriguez JR, Rodriguez D, Heljasvaara R, et al.
(2002) A heterologous prime-boost regime using DNA and recombinant vaccinia
virus expressing the Leishmania infantum P36/LACK antigen protects BALB/c
mice from cutaneous leishmaniasis. Vaccine 20: 1226–1231.
25. Tapia E, Perez-Jimenez E, Lopez-Fuertes L, Gonzalo R, Gherardi MM, et al.
(2003) The combination of DNA vectors expressing IL-12+ IL-18 elicits high
protective immune response against cutaneous leishmaniasis after priming with
DNA-p36/LACK and the cytokines, followed by a booster with a vaccinia virus
recombinant expressing p36/LACK. Microbes Infect 5: 73–84.
26. Abdian N, Gholami E, Zahedifard F, Safaee N, Rafati S (2011) Evaluation of
DNA/DNA and prime-boost vaccination using LPG3 against Leishmania major
infection in susceptible BALB/c mice and its antigenic properties in human
leishmaniasis. Exp Parasitol 127: 627–636.
27. Costa DL, Carregaro V, Lima-Junior DS, Silva NM, Milanezi CM, et al. (2011)
BALB/c mice infected with antimony treatment refractory isolate of Leishmania
braziliensis present severe lesions due to IL-4 production. PLoS Negl Trop Dis 5:
e965.
28. Shi PY, Maizels N, Weiner AM (1997) Recovery of soluble, active recombinant
protein from inclusion bodies. Biotechniques 23: 1036–1038.
29. de Moura TR, Oliveira F, Novais FO, Miranda JC, Clarencio J, et al. (2007)
Enhanced Leishmania braziliensis Infection Following Pre-Exposure to Sandfly
Saliva. PLoS Negl Trop Dis 1: e84.
30. de Moura TR, Novais FO, Oliveira F, Clarencio J, Noronha A, et al. (2005)
Toward a novel experimental model of infection to study American cutaneous
leishmaniasis caused by Leishmania braziliensis. Infect Immun 73: 5827–5834.
31. Titus RG, Marchand M, Boon T, Louis JA (1985) A limiting dilution assay for
quantifying Leishmania major in tissues of infected mice. Parasite Immunol 7:
545–555.
32. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using
real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25:
402–408.
33. de Moura TR, Oliveira F, Rodrigues GC, Carneiro MW, Fukutani KF, et al.
(2010) Immunity to Lutzomyia intermedia saliva modulates the inflammatory
environment induced by Leishmania braziliensis. PLoS Negl Trop Dis 4: e712.
34. Mazumder S, Maji M, Das A, Ali N (2011) Potency, efficacy and durability of
DNA/DNA, DNA/protein and protein/protein based vaccination using gp63
against Leishmania donovani in BALB/c mice. PloS one 6: e14644.
35. Dondji B, Perez-Jimenez E, Goldsmith-Pestana K, Esteban M, McMahon-Pratt
D (2005) Heterologous prime-boost vaccination with the LACK antigen protects
against murine visceral leishmaniasis. Infect Immun 73: 5286–5289.
36. Tewary P, Jain M, Sahani MH, Saxena S, Madhubala R (2005) A heterologous
prime-boost vaccination regimen using ORFF DNA and recombinant ORFF
protein confers protective immunity against experimental visceral leishmaniasis.
J Infect Dis 191: 2130–2137.
37. Rafati S, Zahedifard F, Nazgouee F (2006) Prime-boost vaccination using
cysteine proteinases type I and II of Leishmania infantum confers protective
immunity in murine visceral leishmaniasis. Vaccine 24: 2169–2175.
38. Klinman DM, Kamstrup S, Verthelyi D, Gursel I, Ishii KJ, et al. (2000)
Activation of the innate immune system by CpG oligodeoxynucleotides:
immunoprotective activity and safety. Springer Semin Immunopathol 22:
173–183.
39. Klinman DM, Yamshchikov G, Ishigatsubo Y (1997) Contribution of CpG
motifs to the immunogenicity of DNA vaccines. J Immunol 158: 3635–3639.
40. Carrion J, Folgueira C, Alonso C (2008) Transitory or long-lasting immunity to
Leishmania major infection: the result of immunogenicity and multicomponent
properties of histone DNA vaccines. Vaccine 26: 1155–1165.
41. Andrade BB, de Oliveira CI, Brodskyn CI, Barral A, Barral-Netto M (2007)
Role of sand fly saliva in human and experimental leishmaniasis: current
insights. Scand J Immunol 66: 122–127.
42. Lerner EA, Ribeiro JM, Nelson RJ, Lerner MR (1991) Isolation of maxadilan,
a potent vasodilatory peptide from the salivary glands of the sand fly Lutzomyia
longipalpis. J Biol Chem 266: 11234–11236.
43. Cerna P, Mikes L, Volf P (2002) Salivary gland hyaluronidase in various species
of phlebotomine sand flies (Diptera: psychodidae). Insect Biochem Mol Biol 32:
1691–1697.
44. Charlab R, Valenzuela JG, Rowton ED, Ribeiro JM (1999) Toward an
understanding of the biochemical and pharmacological complexity of the saliva
of a hematophagous sand fly Lutzomyia longipalpis. Proc Natl Acad Sci U S A
96: 15155–15160.
45. Samuelso n J, Lerner E, Tesh R, Titus R (1991) A mouse model of Leishmania
braziliensis braziliensis infection produced by coinjection with sand fly saliva.
J Exp Med 173: 49–54.
46. Donnelly KB, Lima HC, Titus RG (1998) Histologic characterization of
experimental cutaneous leishmaniasis in mice infected with Leishmania
braziliensis in the presence or absence of sand fly vector salivary gland lysate.
J Parasitol 84: 97–103.
47. Lima H, Titus R (1996) Effects of sand fly vector saliva on development of
cutaneous lesions and the immune response to Leishmania braziliensis in BALB/
c mice. Infect Immun 64: 5442.
48. Valenzuel a JG, Belkaid Y, Garfield MK, Mendez S, Kamhawi S, et al. (2001)
Toward a defined anti-Leishmania vaccine targeting vector antigens: charac-
terization of a protective salivary protein. J Exp Med 194: 331–342.
49. Norsworthy N, Sun J, Elnaiem D, Lanzaro G, Soong L (2004) Sand fly saliva
enhances Leishmania amazonensis infection by modulating interleukin-10
production. Infect Immun 72: 1240–1247.
50. Rohousova I, Volf P, Lipoldova M (2005) Modulation of murine cellular
immune response and cytokine production by salivary gland lysate of three sand
fly species. Parasite Immunol 27: 469–473.
51. Gomes R, Teixeira C, Teixeira MJ, Oliveira F, Menezes MJ, et al. (2008)
Immunity to a salivary protein of a sand fly vector protects against the fatal
outcome of visceral leishmaniasis in a hamster model. Proc Natl Acad Sci USA
105: 7845–7850.
52. Tavares NM, Silva RA, Costa DJ, Pitombo MA, Fukutani KF, et al. (2011)
Lutzomyia longipalpis Saliva or Salivary Protein LJM19 Protects against
Leishmania braziliensis and the Saliva of Its Vector, Lutzomyia intermedia.
PLoS Negl Trop Dis 5: e1169.
53. Peters NC, Kimblin N, Secundino N, Kamhawi S, Lawyer P, et al. (2009)
Vector transmission of leishmania abrogates vaccine-induced protective
immunity. PLoS Pathogens 5: e1000484.
54. Goto Y, Bhatia A, Raman VS, Liang H, Mohamath R, et al. (2011) KSAC, the
first defined polyprotein vaccine candidate for visceral leishmaniasis. Clin
Vaccine Immunol 18: 1118–1124.
55. Gomes R, Teixeira C, Oliveira F, Lawyer PG, Elnaiem DE, et al. (2012) KSAC,
a defined Leishmania antigen, plus adjuvant protects against the virulence of L.
major transmitted by its natural vector Phlebotomus duboscqi. PLoS Negl Trop
Dis 6: e1610.
56. Liew FY, Li Y, Millott S (1990) Tumor necrosis factor-alpha synergizes with
IFN-gamma in mediating killing of Leishmania major through the induction of
nitric oxide. J Immunol 145: 4306–4310.
57. Belkaid Y, Hoffmann K, Mendez S, Kamhawi S, Udey M, et al. (2001) The role
of interleukin (IL)-10 in the persistence of Leishmania major in the skin after
healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile
cure. J Exp Med 194: 1497–1506.
58. Falcao S, de Moura TR, Clarencio J, Brodskyn C, Barral A, et al. (2012) The
presence of Tregs does not preclude immunity to reinfection with Leishmania
braziliensis. Int J Parasitol 42: 771–780.
59. Bittencourt A, Silva N, Straatmann A, Nunes VL, Follador I, et al. (2003) Post-
kala-azar dermal leishmaniasis associated with AIDS. Braz J Infect Dis 7: 229–
233.
60. Salay G, Dorta ML, Santos NM, Mortara RA, Brodskyn C, et al. (2007) Testing
of four Leishmania vaccine candidates in a mouse model of infection with
Leishmania (Viannia) braziliensis, the main causative agent of cutaneou s
leishmaniasis in the New World. Clin Vaccine Immunol 14: 1173–1181.
Protective Capacity of Nucleosomal Histones
PLOS ONE | www.plosone.org 9 December 2012 | Volume 7 | Issue 12 | e52296
... Among the proteins predicted as antigenic were ribosomal proteins, DNA/RNA polymerases and kinases that play a role in parasite growth and viability, transcription factors, mitochondrial proteins, nuclear proteins such histones and a considerable number of hypothetical proteins or proteins with unknown function. Importantly, proteins predicted in silico as antigenic in our study, such as beta tubulin, ribosomal proteins and histones, have already been suggested as immunogenic molecules and have already been tested as potential vaccine candidates against visceral as well as cutaneous leishmaniasis [82][83][84][85]. ...
Article
Full-text available
Leishmaniasis is a vector-borne disease caused by an intracellular parasite of the genus Leishmania with different clinical manifestations that affect millions of people worldwide, while the visceral form may be fatal if left untreated. Since the available chemotherapeutic agents are not satisfactory, vaccination emerges as the most promising strategy for confronting leishmaniasis. In the present study, a reverse vaccinology approach was adopted to design a pipeline starting from proteome analysis of three different Leishmania species and ending with the selection of a pool of MHCI- and MHCII-binding epitopes. Epitopes from five parasite proteins were retrieved and fused to construct a multi-epitope chimeric protein, named LeishChim. Immunoinformatics analyses indicated that LeishChim was a stable, non-allergenic and immunogenic protein that could bind strongly onto MHCI and MHCII molecules, suggesting it as a potentially safe and effective vaccine candidate. Preclinical evaluation validated the in silico prediction, since the LeishChim protein, encapsulated simultaneously with monophosphoryl lipid A (MPLA) into poly(D,L-lactide-co-glycolide) (PLGA) nanoparticles, elicited specific cellular immune responses when administered to BALB/c mice. These were characterized by the development of memory CD4+ T cells, as well as IFNγ- and TNFα-producing CD4+ and CD8+ T cells, supporting the potential of LeishChim as a vaccine candidate.
... Hamsters immunized with recombinant histone proteins of L. donovani (rLdh2-4) provided protection against VL (10 7 amastigotes) [330]. BALB/c mice, immunized with plasmids expressing H2A, H2B, H3, and H4 histone proteins, and challenged with L. braziliensis, had lesion development prevented [331]. ...
Article
Full-text available
Leishmaniasis is a zoonotic and vector-borne infectious disease that is caused by the genus Leishmania belonging to the trypanosomatid family. The protozoan parasite has a digenetic life cycle involving a mammalian host and an insect vector. Leishmaniasisis is a worldwide public health problem falling under the neglected tropical disease category, with over 90 endemic countries, and approximately 1 million new cases and 20,000 deaths annually. Leishmania infection can progress toward the development of species–specific pathologic disorders, ranging in severity from self-healing cutaneous lesions to disseminating muco-cutaneous and fatal visceral manifestations. The severity and the outcome of leishmaniasis is determined by the parasite’s antigenic epitope characteristics, the vector physiology, and most importantly, the immune response and immune status of the host. This review examines the nature of host–pathogen interaction in leishmaniasis, innate and adaptive immune responses, and various strategies that have been employed for vaccine development.
... A study by Iborra et al. (2004) evaluated the prophylactic activity of L. infantum histones in an animal model for cutaneous leishmaniasis, and reported that the animals immunized with a mixture of the four plasmids encoding the histones H2A, H2B, H3, and H4 developed a specific Th1 response associated with histone-specific production of IFNγ. Carneiro et al. (2012) also analyzed the immune protection conferred by nucleosomal histones of L. infantum in murine model infected with L. braziliensis, and concluded that histone are potential targets for vaccine formulation against L. braziliensis since they showed significant inhibition activity for the disease. ...
Article
Full-text available
Antigen formulation is the main feature for the success of leishmaniosis diagnosis and vaccination, since the disease is caused by different parasite species that display particularities which determine their pathogenicity and virulence. It is desirable that the antigens are recognized by different antibodies and are immunogenic for almost all Leishmania species. To overcome this problem, we selected six potentially immunogenic peptides derived from Leishmania histones and parasite membrane molecules obtained by phage display or spot synthesis and entrapped in liposome structures. We used these peptides to immunize New Zealand rabbits and determine the immunogenic capacity of the chimeric antigen. The peptides induced the production of antibodies as a humoral immune response against L. braziliensis or L. infantum. Next, to evaluate the innate response to induce cellular activation, macrophages from the peptide mix-immunized rabbits were infected in vitro with L. braziliensis or L. infantum. The peptide mix generated the IFN-γ, IL-12, IL-4 and TGF-β that led to Th1 and Th2 cellular immune responses. Interestingly, this mix of peptides also induced high expression of iNOS. These results suggest that the mix of peptides derived from histone and parasites membrane molecules was able to mimic parasites proteins and induce cytokines important to CD4+ T cell Th1 and Th2 differentiation and effector molecule to control the parasite infection. Finally, this peptide induced an immune balance that is important to prevent immunopathological disorders, inflammatory reactions, and control the parasite infection.
... We build on these currently available tools by developing a dual reporterexpressing L. braziliensis. Besides the advantage of having two stable reporters, this line was developed upon a field isolate (de Moura et al., 2005) that has been widely employed in a variety of studies addressing the pathogenesis of CL caused by L. braziliensis (Novais et al., 2013(Novais et al., , 2017, in addition to drug (Santos et al., 2014;Celes et al., 2016) and vaccine development (Salay et al., 2007;Thalhofer et al., 2011;Carneiro et al., 2012). ...
Article
Full-text available
In this study, we generated a transgenic strain of Leishmania braziliensis, an etiological agent associated with a diversity of clinical manifestations of leishmaniasis ranging from localized cutaneous to mucocutaneous to disseminated disease. Transgenic parasites expressing reporter proteins are valuable tools for studies of parasite biology, host-pathogen interactions, and anti-parasitic drug development. To this end, we constructed an L. braziliensis line stably expressing the reporters eGFP and luciferase (eGFP-LUC L. braziliensis). The integration cassette co-expressing the two reporters was targeted to the ribosomal locus (SSU) of the parasite genome. Transgenic parasites were characterized for their infectivity and stability both in vitro and in vivo. Parasite maintenance in axenic long-term culture in the absence of selective drugs did not alter expression of the two reporters or infection of BALB/c mice, indicating stability of the integrated cassette. Infectivity of eGFP-LUC, L. braziliensis, both in vivo and in vitro was similar to that obtained with the parental wild type strain. The possibility of L. braziliensis tracking and quantification using fluorescence and luminescence broadens the scope of research involving this neglected species, despite its importance in terms of public health concerning the leishmaniasis burden.
... 136 L. infantum histone genes H2A, H2B, H3, and H4 have also been able to control both L. major and L. braziliensis infections in BALB/c mice. 137,138 Similar to the ability to produce chimeric fusion proteins, multiple genes can be fused together for use within third generation vaccines. ...
Article
Full-text available
The leishmaniases are a collection of vector-borne parasitic diseases caused by a number of different Leishmania species that are distributed worldwide. Clinical and laboratory research have together revealed several important immune components that control Leishmania infection and indicate the potential of immunization to prevent leishmaniasis. In this review we introduce previous and ongoing experimental research efforts to develop vaccines against Leishmania species. First, second and third generation vaccine strategies that have been proposed to counter cutaneous and visceral leishmaniasis (CL and VL, respectively) are summarized. One of the major bottlenecks in development is the transition from results in animal model studies to humans, and we highlight that although American tegumentary leishmaniasis (ATL; New World CL) can progress to destructive and disfiguring mucosal lesions, most research has been conducted using mouse models and Old World Leishmania species. We conclude that assessment of vaccine candidates in ATL settings therefore appears merited.
... Histones, as intracellular proteins, are highly conserved among all Leishmania species that can trigger immune responses after spontaneous lysis of Leishmania amastigotes [90,91]. Histone H1 is a highly immunogenic protein that overexpressed in both promastigote and amastigote stages of Leishmania species, especially L. infantum and L. major [92,93]. ...
Article
Visceral leishmaniasis (VL) or kala-azar, the most severe form of the disease, is endemic in more than eighty countries across the world. To date, there is no approved vaccine against VL in the market. Recent advances in reverse vaccinology could be promising approach in designing the efficient vaccine for VL treatment. In this study, an efficient multi-epitope vaccine against Leishmania infantum, the causative agent of VL, was designed using various computational vaccinology methods. Potential immunodominant epitopes were selected from four antigenic proteins, including histone H1, sterol 24-c-methyltransferase (SMT), Leishmania-specific hypothetical protein (LiHy), and Leishmania-specific antigenic protein (LSAP). To enhance vaccine immunogenicity, two resuscitation-promoting factor of Mycobacterium tuberculosis, RpfE and RpfB, were employed as adjuvants. All the aforesaid segments were joined using proper linkers. Homology modeling, followed by refinement and validation was performed to obtain a high-quality 3D structure of designed vaccine. Docking analyses and molecular dynamics (MD) studies indicated vaccine/TLR4 complex was in the stable form during simulation time. In sum, we expect our designed vaccine is able to induce humoral and cellular immune responses against L. infantum, and may be promising medication for VL, after in vitro and in vivo immunological assays.
... For confirmation of results obtained in the RT 2 Profiler PCR arrays, PBMCs from HPs (n = 3) and from LPs (n = 3) were stimulated with L. braziliensis or were cultured in the absence of stimulus (control). RNA was employed in individual quantitative Real Time PCR (qRT-PCR) reactions using primers for IFNG, CXCL10, IFI27, IL6 and IRF1 designed using Primer Express Software (ThermoFisher Scientific), reactions were performed as described elsewhere [10]. qRT-PCRs reactions were run in triplicates for each gene of interest and compared with a housekeeping gene (GAPDH), also using the ΔΔCt method [11]. ...
Article
Full-text available
Background The initial response to Leishmania parasites is essential in determining disease development or resistance. In vitro, a divergent response to Leishmania, characterized by high or low IFN-γ production has been described as a potential tool to predict both vaccine response and disease susceptibility in vivo. Methods and findings We identified uninfected and healthy individuals that were shown to be either high- or low IFN-γ producers (HPs and LPs, respectively) following stimulation of peripheral blood cells with Leishmania braziliensis. Following stimulation, RNA was processed for gene expression analysis using immune gene arrays. Both HPs and LPs were shown to upregulate the expression of CXCL10, IFI27, IL6 and LTA. Genes expressed in HPs only (CCL7, IL8, IFI44L and IL1B) were associated with pathways related to IL17 and TREM 1 signaling. In LPs, uniquely expressed genes (for example IL9, IFI44, IFIT1 and IL2RA) were associated with pathways related to pattern recognition receptors and interferon signaling. We then investigated whether the unique gene expression profiles described here could be recapitulated in vivo, in individuals with active Cutaneous Leishmaniasis or with subclinical infection. Indeed, using a set of six genes (TLR2, JAK2, IFI27, IFIT1, IRF1 and IL6) modulated in HPs and LPs, we could successfully discriminate these two clinical groups. Finally, we demonstrate that these six genes are significantly overexpressed in CL lesions. Conclusion Upon interrogation of the peripheral response of naive individuals with diverging IFN-γ production to L. braziliensis, we identified differences in the innate response to the parasite that are recapitulated in vivo and that discriminate CL patients from individuals presenting a subclinical infection.
Chapter
Full-text available
Visceral and tegumentary leishmaniasis are neglected tropical diseases caused by the protozoan parasite Leishmania. In this chapter, we discuss the causative organisms and the different clinical manifestations, their global and endemic distribution, and methods of vector and human-to-human transmission. We also explore current drug treatment regimens for both diseases and present a brief introduction to vaccine development.KeywordsVisceralTegumentaryLeishmaniasisNeglected tropical diseaseTreatmentDrugsVaccine
Article
Full-text available
Leishmaniasis is an important disease mediated by the protozoan parasite Leishmania via the bite of the female sandfly insect vector. Leishmaniasis is endemic in the tropical and subtropical regions. The most common form of the disease is cutaneous leishmaniasis, which affects more than 10 million people worldwide and includes at least 1.5 million new cases every year. So far, treatment of the disease relies on unsatisfactory chemotherapy that can be complicated by the rising appearance of drug-resistant parasites. Furthermore, it is challenging to achieve solid control of the insect vector and animal reservoir. Therefore, the development of a safe and effective vaccine is urgently needed for the treatment and prevention of leishmaniasis. This review focuses on the recent advances in the development of a safe vaccine that could be used for prevention and treatment of cutaneous leishmaniasis. A short outlook for future research efforts is also presented.
Article
Full-text available
Post-kala-azar dermal leishmaniasis (PKDL) is rarely reported in South America. In spite of the fact that there are many reports about the association of visceral leishmaniasis and AIDS, PKDL is very uncommon in HIV-positive patients, and so far only four cases have been documented in the literature. We present another case with unusual clinicopathological aspects. The patient, a 28-year-old male, from Salvador, Bahia (an endemic area) presented with clinical manifestations of visceral leishmaniasis three years after the diagnosis of AIDS. During treatment for visceral leishmaniasis he developed disseminated miliary papules. Microscopically, the skin biopsy showed a "saw-tooth" pattern with a lichenoid mononuclear infiltrate simulating lichen planus. The histopathological diagnosis was achieved through the finding of amastigotes. The authors discuss the clinicopathological aspects of this case based on a review of the specific literature.
Article
Full-text available
Visceral leishmaniasis (VL) is a fatal disease for humans, and no vaccine is currently available. Sand fly salivary proteins have been associated with protection against cutaneous leishmaniasis. To test whether vector salivary proteins can protect against VL, a hamster model was developed involving intradermal inoculation in the ears of 100,000 Leishmania infantum chagasi parasites together with Lutzomyia longipalpis saliva to mimic natural transmission by sand flies. Hamsters developed classical signs of VL rapidly, culminating in a fatal outcome 5-6 months postinfection. Saliva had no effect on the course of infection in this model. Immunization with 16 DNA plasmids coding for salivary proteins of Lu. longipalpis resulted in the identification of LJM19, a novel 11-kDa protein, that protected hamsters against the fatal outcome of VL. LJM19-immunized hamsters maintained a low parasite load that correlated with an overall high IFN-gamma/TGF-beta ratio and inducible NOS expression in the spleen and liver up to 5 months postinfection. Importantly, a delayed-type hypersensitivity response with high expression of IFN-gamma was also noted in the skin of LJM19-immunized hamsters 48 h after exposure to uninfected sand fly bites. Induction of IFN-gamma at the site of bite could partly explain the protection observed in the viscera of LJM19-immunized hamsters through direct parasite killing and/or priming of anti-Leishmania immunity. We have shown that immunity to a defined salivary protein (LJM19) confers powerful protection against the fatal outcome of a parasitic disease, which reinforces the concept of using components of arthropod saliva in vaccine strategies against vector-borne diseases.
Article
Full-text available
Leishmania parasites are transmitted to their vertebrate hosts by infected phlebotomine sand fly bites. Sand fly saliva is known to enhance Leishmania infection, while immunity to the saliva protects against infection as determined by coinoculation of parasites with vector salivary gland homogenates (SGHs) or by infected sand fly bites (Kamhawi, S., Y. Belkaid, G. Modi, E. Rowton, and D. Sacks. 2000. Science. 290:1351–1354). We have now characterized nine salivary proteins of Phlebotomus papatasi, the vector of Leishmania major. One of these salivary proteins, extracted from SDS gels and having an apparent mol wt of 15 kD, was able to protect vaccinated mice challenged with parasites plus SGH. A DNA vaccine containing the cDNA for the predominant 15-kD protein (named SP15) provided this same protection. Protection lasted at least 3 mo after immunization. The vaccine produced both intense humoral and delayed-type hypersensitivity (DTH) reactions. B cell–deficient mice immunized with the SP15 plasmid vaccine successfully controlled Leishmania infection when injected with Leishmania plus SGH. These results indicate that DTH response against saliva provides most or all of the protective effects of this vaccine and that salivary gland proteins or their cDNAs are viable vaccine targets against leishmaniasis.
Article
Full-text available
The mammalian immune system responds to bacterial infection by rapidly initiating an inflammatory reaction that limits the early spread of the pathogen and facilitates the emergence of antigen-specific immunity (reviewed in [29]). Microorganisms have evolved to avoid such recognition by altering expression of their protein and lipid products [29]. Yet DNA is an indispensable and highly conserved component of all bacteria. Indeed, the genomes of otherwise diverse prokaryotes share DNA motifs that are rare in the genomes of higher vertebrates [3, 11, 30]. Yamamoto et al. [50, 51] were the first to show that bacterial but not mammalian DNA boosted the lytic activity of NK cells, inducing them to secrete IFN-γ. They attributed this effect to palindromic sequences present in bacterial DNA [51]. Other investigators showed that bacterial DNA stimulated B cells to proliferate and secrete Ig (especially if the DNA was complexed to a DNA-binding protein) [10–12, 30, 34, 49]. We were interested in defining the precise sequence motifs in bacterial DNA responsible for this immune activation, and to establish the breadth and nature of such activation.
Article
Abstract Leishmaniases are wide spread diseases transmitted to their vertebrate host by infected sand fly. The saliva from these arthropods contains a vast repertoire of pharmacologically active molecules that hampers the host’s haemostatic, inflammatory and immune responses. The early interactions between Leishmania and the host’s immune response are closely linked to disease evolution or protection against the protozoan, and the ectoparasite saliva contributes directly to these interactions. Current studies have depicted these features, and these relations are being widely explored. There are concrete indications that the host response against sand fly saliva influences disease outcome in leishmaniasis. Additionally, there are demonstrations that immunization with whole sand fly saliva, or its components, leads to protection against leishmaniasis in different host species. The combination of these evidences opens up optimistic perspectives for improving vaccine development against Leishmania infection.
Article
The two most commonly used methods to analyze data from real-time, quantitative PCR experiments are absolute quantification and relative quantification. Absolute quantification determines the input copy number, usually by relating the PCR signal to a standard curve. Relative quantification relates the PCR signal of the target transcript in a treatment group to that of another sample such as an untreated control. The 2(-DeltaDeltaCr) method is a convenient way to analyze the relative changes in gene expression from real-time quantitative PCR experiments. The purpose of this report is to present the derivation, assumptions, and applications of the 2(-DeltaDeltaCr) method. In addition, we present the derivation and applications of two variations of the 2(-DeltaDeltaCr) method that may be useful in the analysis of real-time, quantitative PCR data. (C) 2001 Elsevier science.
Article
Leishmania spp. cause a broad spectrum of diseases collectively known as leishmaniasis. Leishmania braziliensis is the main etiological agent of American cutaneous leishmaniasis and mucocutaneous leishmaniasis. During experimental infection with L. braziliensis, BALB/c mice develop an adaptive immune response that is associated with lesion healing and, in parallel, parasite persistence within draining lymph nodes (dLNs). In the Leishmania major model of cutaneous leishmaniasis, regulatory T cells (Tregs) play an important role in immune regulation, preventing pathological immune responses but at the same time precluding sterile cure. In this study we investigated the role of Tregs during experimental infection with L. braziliensis. CD4(+)CD25(+) T cells were detected throughout the duration of clinical disease both at the ear and in dLNs of infected mice. These cells expressed Treg markers such as glucocorticoid-induced TNF-receptor-related protein (GITR), the α chain of the αεβ7 integrin (CD103), and the forkhead/winged helix transcription factor, Foxp3, and were able to suppress the proliferation of CD4(+)CD25(-) cells. Importantly, a high frequency of Foxp3(+) cells accumulated at the site of infection and in dLNs. We next investigated the outcome of a reinfection with L. braziliensis in terms of Treg distribution and disease reactivation. Interestingly, a secondary inoculation with L. braziliensis did not preclude an efficient recall response to L. braziliensis at a distal site, despite the presence of Tregs. Within dLNs, reinfection did not promote parasite dissemination or a differential recruitment of either CD4(+)CD25(+)Foxp3(+) or CD4(+)IL-10(+) T cells. On the contrary, parasites were mainly detected in the LN draining the primary infection site where a high frequency of CD4(+)IFN-γ(+) T cells was also present. Collectively these data show that during experimental infection, Tregs are present in healed mice but this population does not compromise an effective immune response upon reinfection with L. braziliensis.