You are on page 1of 15

Journal of Proteomics 232 (2021) 104077

Contents lists available at ScienceDirect

Journal of Proteomics
journal homepage: www.elsevier.com/locate/jprot

Quantitative analysis of proteins secreted by Leishmania (Viannia)


braziliensis strains associated to distinct clinical manifestations of American
Tegumentary Leishmaniasis
Andrés Rodríguez-Vega a, 1, 2, Monica Losada-Barragán b, 2, Luiz Ricardo Berbert c, 3,
Camila Mesquita-Rodrigues d, Ana Cristina Souza Bombaça e, Rubem Menna-Barreto e,
Priscila Aquino f, Paulo C. Carvalho g, Gabriel Padrón a, 4, Jose Batista de Jesus d, h,
Patricia Cuervo a, *
a
Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brazil
b
Grupo de Investigación en Biología Celular y Funcional e Ingeniería de Biomoléculas, Universidad Antonio Nariño, Bogotá, Colombia
c
Laboratório de Pesquisas sobre o Timo, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brazil
d
Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brazil
e
Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brazil
f
Instituto Leônidas e Maria Deane, Fiocruz, Manaus, AM, Brazil
g
Laboratory for Structural and Computational Proteomics, Fiocruz-Paraná, PR, Brazil
h
Universidade Federal de São João Del Rei, São João del Rei, MG, Brazil

A R T I C L E I N F O A B S T R A C T

Keywords: The role of Leishmania braziliensis in the development of different clinical forms of American Tegumentary
Leishmania braziliensis Leishmaniasis (ATL) is unclear, but it has been suggested that molecules secreted/released by parasites could
Secretome modulate the clinical outcome. Here, we analyzed the infection rate and cytokine profile of macrophages pre­
Localized cutaneous leishmaniasis
treated with the secretome of two L. braziliensis strains associated with polar clinical forms of ATL: one associated
Disseminated cutaneous leishmaniasis
Quantitative proteomics
with localized self-healing cutaneous leishmaniasis (LCL) and other associated with the disseminated form (DL).
Mass spectrometry Besides, we use an iTRAQ-based quantitative proteomics approach to compare the abundance of proteins
secreted by those strains. In vitro infection demonstrated that pretreatment with secretome resulted in higher
number of infected macrophages, as well as higher number of amastigotes per cell. Additionally, macrophages
pretreated with LCL secretome exhibited a proinflammatory profile, whereas those pretreated with the DL one
did not. These findings suggest that secretomes made macrophages more susceptible to infection and that
molecules secreted by each strain modulate, differentially, the macrophages’ cytokine profile. Indeed, prote­
omics analysis showed that the DL secretome is rich in molecules involved in macrophage deactivation, while is
poor in proteins that activate proinflammatory pathways. Together, our results reveal new molecules that may
contribute to the infection, persistence and dissemination of the parasite.
Significance: Leishmania braziliensis is associated to localized self-healing cutaneous lesions (LCL), disseminated
leishmaniasis (DL), and mucocutaneous lesions (MCL). To understand the role of the parasite in those distinct
clinical manifestations we evaluated infection rates and cytokine profiles of macrophages pre-treated with
secretomes of two L. braziliensis strains associated with DL and LCL, and quantitatively compared these secre­
tomes. The infection index of macrophages pretreated with the DL secretome was significantly higher than that
exhibited by non-treated cells. Interestingly, whereas the LCL secretome stimulated a proinflammatory setting,

* Corresponding author at: Laboratório de Pesquisa em Leishmanioses, Instituto Oswaldo Cruz, FIOCRUZ. Av. Brasil 4365, Manguinhos, 21040-360, Rio de Janeiro,
RJ, Brazil.
E-mail address: patricia.cuervo@fiocruz.br (P. Cuervo).
1
Current Address: Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico,
Universidade do Estado do Rio de Janeiro.
2
These authors contributed equally to this work.
3
Current Address: Coordenação de Atividades com Modelos Biológicos Experimentais - Decania do Centro de Ciências da Saúde - UFRJ
4
Current Address: Center for Genetic Engineering & Biotechnology, La Habana, Cuba

https://doi.org/10.1016/j.jprot.2020.104077
Received 31 August 2020; Received in revised form 1 December 2020; Accepted 6 December 2020
Available online 10 December 2020
1874-3919/© 2020 Elsevier B.V. All rights reserved.
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

favoring an effector cell response that would explain the proper resolution of the disease caused by this strain,
the DL strain was not able to elicit such response or has mechanisms to prevent this activation. Indeed, DL
secretome is rich in peptidases that may deactivate cell pathways crucial for parasite elimination, while is poor in
proteins that could activate proinflammatory pathways, favoring parasite infection and persistence.

1. Introduction and other professional phagocytic cells to the infection site. Once the
parasites come into contact with the phagocytic cells and/or are inter­
The American tegumentary leishmaniasis (ATL) comprises a group of nalized, they secrete several molecules that can modulate the activation
diseases caused by multiple Leishmania species and with a broad spec­ of the macrophage, subverting the beginning of an appropriate response
trum of clinical manifestations and therapy response [1,2]. Near one to the infection [26,27].
million cases of ATL were reported in the Americas during the last two Based on those evidence, we hypothesize that proteins secreted by
decades, with an average of ~55,000 cases every year [3,4]. L. braziliensis specifically during the initial process of macrophages
Leishmania (Viannia) braziliensis is one of the main species respon­ infection may modulate the activation profile of the cells, contributing
sible for ATL in the continent. This species is associated with different to the disparity in the clinical outcome of infections with this species. In
clinical forms ranging from localized self-healing cutaneous lesions order to evaluate this, here we studied the secretomes of two different L.
(localized cutaneous leishmaniasis, LCL) to multiple cutaneous lesions braziliensis strains: one isolated from a patient with LCL and one isolated
and/or oropharyngeal mucosa compromise. The persistence of the from a patient with DL. We evaluated the effect of the secretomes of each
parasite after the appearance of the single initial lesion and its capability strain on the infection rate and cytokine profile of peritoneal macro­
to metastasize, are remarks of the infection with this species. Lymphatic phages in vitro, as well as using an iTRAQ-based quantitative proteomics
or haemotogenous metastatic dissemination of parasites may result in approach we identified and compared the relative abundance of proteins
lesions in the oropharyngeal mucosa and cartilage (mucocutaneous secreted by each strain. Our data provide evidence on the differential
leishmaniasis, MCL), which occurs in 1–10% of infections, or may result infection rate and inflammatory profile of macrophages pre-stimulated
in the appearance of multiple skin lesions (disseminated leishmaniasis, with the secretion of each strain. In addition, we describe the proteins
DL), frequently refractory to treatment [5–9]. Remarkable, cutaneous in those secretomes, their relative abundance, and the main quantitative
and mucocutaneous lesions can lead to cosmetic disfiguration with differences between those strains.
consequent social stigmatization and psychological sequels [10,11]. In
the severe forms of MCL, it can lead to the destruction of the nasal 2. Materials and methods
septum and palate, resulting in a highly, life-threatening, disfiguring
condition. Disseminated leishmaniasis is characterized by ten or more 2.1. Ethics statement
cutaneous lesions located in two or more parts of the body, few parasites
in the lesions, and a high frequency of mucosal involvement [12–18]. This study was carried out in accordance with the recommendations
The clinical pleiomorphism observed in infections with L.braziliensis of the Guide for the Care and Use of Laboratory Animals of the National
results from the complex interaction of particular features of the para­ Institutes of Health (NIH Publications No. 8023, revised 1978). All an­
sites and host’ immune response. While proinflammatory cytokines such imal procedures were approved by the Instituto Oswaldo Cruz Animal
as TNF-α and IFN-γ are associated with lesion resolution in the case of Care and Use Committee (License # L-005/2017).
localized CL, high levels of those cytokines lead to the destruction of
mucosal tissue in the case of MCL and systemic decreased levels of the 2.2. Parasite culture
same cytokines seem to be associated with DL, indicating a weak
response mediated by T cells that leads to a failure in the control of The L.braziliensis strains used in this study were provided by the
parasites but favors little tissue destruction [15,17,19]. Nevertheless, Collection of Leishmania of the Instituto Oswaldo Cruz (Coleção de
the mechanisms involved in the progression of the skin lesion to MCL or Leishmania do Instituto Oswaldo Cruz, CLIOC, http://clioc.fiocruz.br/).
DL forms are still not well understood. How does L. braziliensis metas­ The strain MHOM/BR/2000/LTCP13396 associated with DL and the
tasize? How is it explained that only in some infections does the parasite strain MHOM/BR/2000/LTCP13490 associated with LCL were isolated
disseminate? The parasite’s role in the dissemination process is not yet in the same geographical region, belong to the same zymodema, had
clear, but it is suggested that the intrinsic characteristics of some strains their in vitro passages controlled and their infectious capacity main­
may modulate the clinical outcome. Studies in a L. braziliensis-rhesus tained through inoculation in golden hamsters. As mentioned above,
model showed that infection with a strain isolated from an LCL patient such strains reproduced the clinical phenotype in the rhesus model
resulted in self-resolution [20], whereas infection with a strain from a [20,21]. Through the text, tables, and figures, those strains will be
DL patient caused multiple disseminated lesions in 100% of the monkeys mentioned as DL and LCL. Promastigotes were cultivated at 25 ◦ C in
[21]. These observations suggest that the parasites that disseminated biphasic NNN- Schneider’s medium (Vitrocell) supplemented with 20%
can be distinguished from those that do not and that such strains, fetal bovine serum (FBS) (Vitrocell, heat-inactivated at 56 ◦ C for 50 min)
associated with very polar clinical manifestations [20,21], represent a at 25 ◦ C. According to the Brazilian Law of Biodiversity, this study was
good model for studying the parasite’ factors involved in dissemination registered at SisGen (AA2236F).
and clinical outcome.
Secreted molecules, in particular proteins, are key mediators of
2.3. Secretion assays
communication between Leishmania and host cells and participate in
several physiological processes, such as cell signaling, differentiation,
Secretion assays were performed as previously described [25], with
invasion, dissemination, among others [22–24]. Indeed, during the
minor modifications. Briefly, 5 × 108 promastigotes were centrifugated
description of the L. braziliensis secretome, our group identified several
and washed twice with PBS (0.01 M phosphate-buffered 0.145 M NaCl,
virulence factors and molecules involved in immunomodulation, rein­
pH 7.2). Parasites were resuspended in 5 mL of RPMI 1640 medium
forcing the idea that inherent features of parasites could have a role in
(Gibco) supplemented with 25 mM HEPES and incubated for 3 h at
the clinical outcome [25]. Accordingly, studies of different Leishmania
25 ◦ C. Before used, RPMI supplemented medium was tested for endo­
spp. showed that proteins secreted in the first moments of the infection
toxin using the commercial kit endosafe (Charles River Labs). Subse­
have an essential role in the recruitment of neutrophils, macrophages,
quently, supernatants were collected by centrifugation at 1800 xg for 10

2
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

min for parasites removal followed by filtration through a 0.2 μm filter with iTRAQ 4-plex (AB Sciex, Framingham, MA, USA) according to the
unit (Millipore, Bedford, MA) for debris removal. The conditioned me­ instruction of the manufacturer. Labeling proceeded as follows: two
dium so obtained contains the secretome that was used in further ex­ biological replicates of DL secretomes were labeled with iTRAQ114 and
periments. The assays were carried out in biological quadruplicates. two were labeled with iTRAQ116, whereas two biological replicates of
During the secretion assay, parasites’ viability was evaluated every LCL secretomes were labeled with iTRAQ115 and two with iTRAQ117.
hour by flow cytometry using propidium iodide (PI) labeling. For posi­ Labeled peptides were mixed, pooling one sample of each label, total­
tive cell death control, parasites were permeabilized with 1% BSA and izing two final mixes. Both pools were fractionated by strong cation
0.1% saponin for 15 min at 25 ◦ C. Samples were incubated with 2 μL of exchange using Macro Spin Columns (Harvard Apparatus, USA), and the
PI (1 mg/mL) at 25 ◦ C for 20 min protected from light. Acquisition two resulting fractions were dried in a vacuum centrifuge system and
(10,000 events) was performed on a FACSCanto™ flow cytometer desalted using Spin Column C18 (Harvard Apparatus, USA).
(Becton Dickinson, USA). Off-line analysis was made using the Sum­
mit™ software. 2.7. LC-MS/MS analysis

2.4. Peritoneal macrophages infection and treatment with secretomes Samples were analyzed by nano-liquid chromatography (Easy-nLC
1000, Thermo Fischer Scientific) coupled to tandem mass spectrometry
To evaluate whether the secretome of each L.braziliensis strain could (nLC-MS/MS) in the mass spectrometry facility of Carlos Chagas Insti­
modulate the percentage of infection of macrophages in vitro, murine tute - Fiocruz Paraná (PDTIS-RPT02H). Samples were injected in
peritoneal macrophages were pre-stimulated with secretomes obtained duplicate. Peptides were loaded onto a reverse phase column (30 cm
as described above, infected, and further analyzed by microscopy. long, 75 μm inner diameter) packed in-house using a flow of 500 nL/min
Macrophages were obtained by peritoneal washes from Swiss mice, ReprosilPur C18 Acqua (beads 1.9 μm in diameter, Dr. Maisch). Peptide
resuspended in RPMI 1640 supplemented with 10% SFB, 4 mM L- elution followed a flowrate of 250 nL/min, and a gradient of 5–40% of
glutamine, 1000 U/mL penicillin and 50 μg/mL streptomycin, and phase B (5% DMSO, 0.1% formic acid in acetonitrile) for 180 min. Phase
grown on plates containing sterile glass coverslips for 24 h at 37 ◦ C and A was 5% DMSO in 0.1% formic acid. Data were acquired in data-
5% CO2. Further, cells were stimulated for 24 h with 0.6 μg of the dependent acquisition mode, automatically switching between full
endotoxin-free secretome from DL or LCL strain, which corresponds to scan MS (m/z 300–2000) at a resolution of 60,000 (m/z 100) and MS/
the quantity of protein secreted by 106 parasites, resembling a MOI of MS acquisition, with a dynamic exclusion of 90 s in the LTQ Orbitrap XL-
10:1 (parasite:macrophage). Control macrophages were incubated with ETD mass spectrometer (Thermo Fisher Scientific). For each MS spec­
supplemented RPMI for 24 h. After stimulation, the conditioned medium trum, up to the five more intense ions with +2 and + 3 charges were
was removed and macrophages (control and treated) were infected with isolated and fragmented through high energy collision dissociation
DL or LCL strain at a MOI of 10:1 and incubated for 3 h. Infected mac­ (HCD) with a normalized collision energy of 45 and a 30 ms activation
rophages were further washed to remove non-internalized promasti­ time. Xcalibur 2.0 software (Thermo Fischer Scientific) was used to
gotes and incubated for 24, 48, and 72 h. Cell-free supernatants were control the scan functions and solvent gradients in the nLC.
collected at the indicated times and stored at − 80 ◦ C for further cytokine
quantitation. For calculation of the infection rate, coverslips were 2.8. Protein identification and quantification
stained with panoptic staining, and cells were examined by light mi­
croscopy (Nikon Eclipse E400-Tokyo, Japan) (60× objective). The Proteins were identified using the software PatternLab for Prote­
number of infected cells, as well as the number of amastigotes per cell, omics (V.3.2.0.3) (http://patternlabforproteomics.org/) and a custom­
were assessed. Assays were performed in triplicate. ized database containing protein sequences available for Leishmania spp.
at UniProt in January 2017 (http://www.uniprot.org/, [29]). Peptide
2.5. Cytokine quantitation and protein false identification rate (FDR) was set at 1% based on the
target-decoy reverse database, including also common contaminants
Supernatants collected from peritoneal macrophages at indicated available in the PatternLab’s Search Database Generator [30]. Search
times were analyzed by flow cytometry using a mouse CBA inflamma­ parameters used for identification were: tryptic and semi-tryptic pep­
tion kit (Cytometric Beads Array, Cat No. 552364, Becton Dickinson, tides; three missed cleavages; fixed modification of cysteine carbami­
USA) for quantification of IL-6, IL-10, MCP-1/CCL2, IFN-γ, TNF-α, and domethylation; fixed iTRAQ (+144.1 Da) at the N-terminal; variable
IL-12p70. Samples were prepared according to the manufacturer’s in­ iTRAQ modification (+144.1 Da) in lysine (K) side chain; and 40 ppm
structions and acquired on a FACSCanto™ flow cytometer (Becton mass tolerance of the precursor. Peptide-spectrum matches (PSM) were
Dickinson, USA). Data were analyzed using FCAP Array Software v3.0 ™ validated with the Search Engine Processor (SEPro), in PatternLab’s
software. environment [30]. Identifications were grouped by charge state (2+ and
3+) and then by a fully tryptic or semi-tryptic state. Six amino acid
2.6. ITRAQ-based protein quantification residues were set as a minimum for sequence length. A bayesian
discriminator for each result was produced based on the ProLuCID
Secretomes were concentrated to a final volume of 100 μL using XCorr, DeltaCN, and ZScore values. In addition, results with two or more
centrifugation filters 3 K (Millipore), and proteins were precipitated evidence of the protein in the sample and sequences with less than five
with trichloroacetic acid (15% v/v) for 2 h at 4 ◦ C and washed twice with ppm of variation at the protein level were accepted. Identified proteins
cold acetone. Proteins were resuspended in triethylammonium bicar­ in the database corresponding to L. braziliensis species, as well as some
bonate buffer (TEAB) 50 mM pH 8.5, dosed using a Qubit® fluorometer other Leishmania species, were accepted and cured manually.
(Invitrogen) and processed as described previously [28]. Briefly, pro­ Relative quantification of proteins was made as described previously
teins were treated with 0.1% of Rapigest (Waters), reduced by the [28], using the ‘Isobaric Analyzer’ module of PatternLab [31,32]. A two-
addition of 20 mM tris(2-carboxyethyl)-phosphine (TCEP) at 60 ◦ C for class analysis was made using the quantified peptides for comparing DL
30 min, alkylated with iodoacetamide (IAA) 20 mM at 25 ◦ C for 30 min. strain (iTRAQ114 and iTRAQ116) to LCL strain (iTRAQ115 and
Afterward, samples were digested sequentially with lysyl endopeptidase iTRAQ117 labels). A cutoff of at least two unique peptides per protein,
mass spectrometry grade (Wako) and sequencing grade trypsin (Prom­ 0.30 of “peptide Log Fold Change Cutoff” and 0.05 of “Peptide p-value
ega). Peptides were desalted in a spin column C18 (Harvard Apparatus, Cutoff” for the t-paired test or p binomial value was set. p-value was
USA), dried by vacuum centrifugation and resuspended in TEAB 50 mM. calculated according to the Benjamini-Hochberg procedure [32]. Pro­
Four biological replicates of secretomes of each strain were labeled teins with a fold change of abundance ≥1.5 and p ≤ 0.05 were accepted.

3
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Identified proteins were classified according to cellular component, displayed viability above 97% after 1, 2, and 3 h of secretion (Supple­
molecular function, and biological process through Gene Ontology mentary Fig. 1).
database (http://geneontology.org/) using the Gene Ontology Explorer To assess whether the secretome from DL or LCL strains could
tool (GOEx) from the PatternLab [31]. Interactome network analysis differentially influence the infection rate, peritoneal macrophages were
was built using the web-accessible program IIS- Integrated Interactome treated with the secretome of each strain and subsequently were infec­
system (http://www.lge.ibi.unicamp.br/lnbio/IIS/) [33]. The enrich­ ted. The treatment with parasite secretome, DL or LCL, before challenge
ment level of proteins with any annotation of the biological process was with DL or LCL parasites significantly increased the number of infected
calculated using the orthologous coding genes in humans. Data were cells at all analyzed times for both strains (Fig. 1A). Also, the number of
visualized using Cytoscape version 2.8.3 (http://www.cytoscape.org/). parasites per cell at 24 and 72 h post-infection with DL strain, and at 48 h
post-infection with LCL strain, was significantly increased when mac­
2.9. Bioinformatics analysis of secreted proteins rophages were pretreated with secretion (Fig. 1B). Remarkably, the
infection index of macrophages pretreated with the DL secretome was
Different servers and databases were used to analyze functional do­ significantly higher than that exhibited by non-treated macrophages in
mains, secretion pathways, and exosomal origin of the identified pro­ all the times evaluated (Fig. 1C). For macrophages pretreated with LCL
teins. Prediction of functional domains for uncharacterized proteins was secretome, the infection index was higher than for non-treated ones only
made using Pfam V27.0 (http://pfam.xfam.org/) [34]. SignalP 4.1 (http at 48 h.
://www.cbs.dtu.dk/services/SignalP-4.1/) was used for the prediction
of N-terminal signal peptide [35]. Potential non-classical secretion 3.2. LCL secretome and infection stimulated a proinflammatory cytokine
pathways and potential subcellular location were predicted with profile in peritoneal macrophages in vitro whereas DL secretome did not
SecretomeP 2.0 (http://www.cbs.dtu.dk/services/SecretomeP/) [36]
and TargetP 1.1 (http://www.cbs.dtu.dk/services/TargetP/) [37], As we observed that the infection index of peritoneal macrophages
respectively. The TMHMM v.2.0 server (http://www.cbs.dtu.dk/servic was significantly increased when the cells were pretreated with the
es/TMHMM/) [38] was used for the prediction of transmembrane do­ secretome of DL strain, we decided to evaluate the cytokine profile of
mains. Finally, our dataset was compared against the ExoCarta database those macrophages and compare it to the profile of macrophages stim­
(http://www.exocarta.org/) [39] to determine the potential exosomal ulated with LCL secretome. We observed significant differences in the
origin of the identified proteins. levels of cytokines produced by pretreated-infected macrophages
compared to untreated-infected ones, as well as significant differences
2.10. Gene expression analysis between the cytokine profiles induced by each secretome (DL or LCL).
Macrophages pre-stimulated with LCL secretome and further infec­
To assess whether the differential protein abundance in the secre­ ted with that strain showed significantly higher levels of TNF-α and IL-6
tome could correlate with the relative abundance of their transcripts, a (p < 0.05) compared with the levels measured for macrophages pre­
gene expression analysis was performed for six genes of both strains treated and infected with DL strain, at all analyzed times (Fig. 2). TNF-α
using quantitative real-time PCR (qPCR) following protocols previously and IL-6 levels were 4 to 8-fold higher in the LCL pretreated-infected
described [40]. Total RNA was extracted from promastigotes using cells than in the DL pretreated-infected macrophages (Fig. 2). Interest­
Trizol; RNA was quantified, and the cDNA was synthesized using the ingly, higher levels of those cytokines were also observed when unsti­
SuperScript III reverse transcription system (Invitrogen). qPCR was mulated macrophages were infected with LCL strain; however, the TNF-
performed using a StepOne equipment and SYBR® Green PCR Master α and IL-6 concentrations were 2-fold lower than those observed in pre-
Mix (Applied Biosystems). Oligonucleotide sequences of the target and stimulated macrophages (Figs. 2 and 3). In addition, at 72 h post-
reference genes were designed from available public sequences using infection, the concentration of CCL2 was significantly higher (3-fold)
Primer Blast software (Supplementary Table 1). PCR consisted of 40 in macrophages pre-treated and infected with LCL strain than in those
cycles of 10 s at 95 ◦ C, 15 s at 56 ◦ C and 10 s at 72 ◦ C, followed by stimulated with DL secretome and infected (Fig. 2, p < 0.001). The
melting curve analyzes in order to verify the reaction specificity. Stan­ concentration of CCL2 was also significantly higher in untreated mac­
dard curves for all genes were generated using serial cDNA dilutions rophages infected with LCL than in cells infected with DL strain (Fig. 3).
obtained from all samples. Gene expression was quantified using the Remarkably, although IL-12 and IL-10 were detected at 24 h and 48 h
comparative Ct method (ΔΔCt). Data are shown as normalized re­ in macrophages pre-stimulated with DL secretome and infected, the
lationships between target gene expression and geometric mean refer­ levels of those cytokines were abrogated at 72 h post-infection. More­
ence genes [41]. The experiments followed the Minimum Information over, IL-12 and IFN-γ levels were almost undetectable in cells pre-
for Publication of Quantitative Real-Time PCR Experiments (MIQE) treated and infected with LCL strain (Fig. 2) at all times evaluated
guidelines [41,42]. here, whereas IL-10 was significantly higher in those cells, at 72 h post-
infection, when compared to macrophages pre-treated and infected with
2.11. Statistical analysis DL strain. In contrast, unstimulated macrophages infected with LCL
strain displayed detectable levels of IL-12, IL-10, and IFN-γ at all
Statistical analysis was performed using GraphPad Prism 5.0 soft­ analyzed times. In contrast, those cytokines were barely detected in
ware. To analyze the differences between the two conditions studied, untreated macrophages infected with DL strain (Fig. 3).
Student’s t-test was applied. Data are presented as means ± standard
deviation. 3.2.1. Secretome contains proteins secreted mainly by alternative pathways
involving exosomes
3. Results As the secretomes of the LCL and DL strains differentially altered the
infection rate and cytokine profile of macrophages pre-treated with
3.1. The secretome of L. braziliensis increases the infection rate of them, we hypothesized that the proteins secreted by each strain can
peritoneal macrophages in vitro differentially modulate the host’s immune response, leading to polarized
clinical manifestations. Therefore, we applied iTRAQ labeling to identify
First, to ensure that during the 3 h of secretion assays the proteins and quantify the differences in protein abundance between LCL and DL
found in the conditioned medium are derived from active secretion secretomes. A total of 1176 peptides identified were assigned to 252
rather than released by cell death, parasite viability was measured every proteins at 1% FDR (Supplementary Table 2). From these proteins, 80%
hour by flow cytometry through PI labeling. Both DL and LCL strains (201/252) were identified with two peptides at least, while 20% (51/

4
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Fig. 1. Effect of pre-treatment with L. (V.) braziliensis secretome on in vitro macrophage infection. Murine peritoneal macrophages were pre-treated with secretion of
each L. (V.) braziliensis strain and infected with promastigotes of the corresponding strain. Infection was followed at 24, 48 and 72 h post infection. (A) Percentage of
infected cells; (B) number of parasites per macrophage; (C) infection index. Statistical differences between control and pretreated cells were analyzed by Student’s t-
test (*p < 0.05, **p < 0.005 and ***p < 0.001) (n = 3). Graphs represent mean ± standard deviation.

252) were identified with a single peptide (Supplementary Table 2). secreted by classical or alternative pathways according to the pre­
Proteins identified with only one peptide were accepted since there was dictions obtained by SignalP, SecretomeP and/or exocarta, suggesting
more than one evidence of their presence in the sample, such as two or that they could be truly exported to the extracellular milieu. The other
more spectra and two or more charge states. ~3% proteins could be part of apoptotic vesicles or shedding products,
Bona fide secreted proteins were identified in the L. braziliensis known to be part of the parasite secretion and important for the infective
secretome, including GP63, secreted acid phosphatase, disulfide isom­ process [24]. Finally, for ~20% proteins, it was not possible to predict
erase protein, and activated protein kinase c receptor, among others any signal of secretion with the tools used here, but several of them have
(Supplementary Table 2). Bioinformatics analyses showed that ~80% been found enriched in the conditioned medium of several Leishmania
identified proteins could be secreted by either classical or alternatives spp. [24,25,43].
routes, and for several proteins, one or more pathways were predicted To obtain information about the potential function of the unchar­
(Supplementary Table 2 and Fig. 4). In addition, those predictions acterized proteins, we analyzed functional domains using the Pfam 27.0
showed that the predominant pathway of secretion involves exosomes server. We found at least one functional domain for some regions of the
(Supplementary Table 2 and Fig. 4). TMHMM analysis showed that ~9% proteins in ~45% of the cases (25/56). Among these, we highlight heat
of identified proteins contain at least one transmembrane domain shock proteins, thioredoxin, and phosphatase domains, among others
(Supplementary Table 2). However, ~6% of those proteins also can be (Supplementary Table 3).

5
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Fig. 2. Cytokines secreted by murine macrophages pre-treated with secretome of the DL or LCL strain of L. (V.) braziliensis and infected. Cytokines were quantified by
flow cytometry in supernatants of macrophages pre-treated with the secretome of each strain and infected with the homologous strain. Statistically significant
differences between control and treatment were calculated using the Student’s t-test, using the GraphPad Prism 5.0 program. (*) p < 0.04 and (**) p < 0.008; (***) p
< 0.0009. The bars represent mean ± standard deviation.

3.2.2. Proteins secreted by L. braziliensis are mainly involved in catalytic redox metabolism such as SOD, trypanothione reductase, and trypar­
activities edoxin peroxidase, among others (Table 1), and in the case of proteins
In order to obtain more information about the function of identified involved in protein degradation, 17 were identified, including peptidases
proteins, we retrieved their annotations regarding the main gene recognized for their role in virulence, such as GP63 and calpain-like
ontology categories: biological process, molecular function, and cellular cysteine peptidases (Table 1).
component (Supplementary Fig. 2). We observed that 71% (180/252)
proteins have an annotation in at least one of the three categories, while 3.3. LCL and DL associated strains show significant differences in protein
~29% (72/252) of the proteins did not have an associated classification. abundance in their secretomes
According to the molecular function annotations, the subcategories
associated with catalytic activity (44%) or binding (40%) were more To gain insights into the quantitative differences of protein abun­
represented in our dataset. Ninety-four proteins were classified into six dance between the two strains of L. braziliensis studied here, we used a
different categories: (i) stress response and protein folding, (ii) carbohy­ combination of stable isotope labeling with tandem mass spectrometry
drate metabolism, (iii) amino acid metabolism, (iv) nucleotide metabolism, to compare the relative abundance of protein in the secretome of LCL
(v) protein degradation and (vi) protein translation and synthesis. and DL strains. Differences in the abundance of secreted proteins be­
Furthermore, the ontological classification allowed us to focus on cate­ tween the strains were calculated in the Quant-Isobaric analyzer module
gories of interest for the study of the first interactions of the parasite of PatternLab by comparing the iTRAQ normalized reporter ion in­
with host cells. In response to stress and protein folding, for example, 18 tensities. A total of 26 proteins showed significant differences in abun­
proteins were found, including heat shock proteins and proteins of the dance between the strains (Table 2). The relative abundance of 13

6
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Fig. 3. Cytokines secreted by murine macrophages infected with DL or LCL strain of L. (V.) braziliensis. Cytokines were quantified by flow cytometry in macrophages
infected with the DL or LCL strain. Statistically significant differences between control and treatment were calculated using the Student’s t-test, using the GraphPad
Prism 5.0 program. (*) p < 0.03; (**) p < 0.006 and (***) p < 0.0009. The bars represent mean ± standard deviation.

proteins was increased in the DL strain compared to the LCL strain, in parasite differentiation process as well as in binding to RNA and
whereas the relative abundance of 13 proteins was decreased (Table 2). regulation of virulence factors [44,45].
Interestingly, among the proteins with increased abundance in the As the canonical function described for various of those proteins do
secretome of the DL strain, there are several proteins involved in the not allow understanding their role in the extracellular milieu, we
response to stress and redox homeostasis, such as the thioredoxin-like_fold analyzed if those proteins could have an alternative function previously
domain containing protein and a trypanothione reductase. We also described. Remarkably, four proteins that were found differentially
observed that proteins involved in protein degradation and cell adhesion abundant between the strains showed similarity with multitasking
such as the dipeptidyl-peptidase 3 and the uncharacterized protein proteins described in other organisms (Supplementary Table 4). Dihy­
A4H3T9, as well as in translation and protein synthesis such as the 40S drolipoyl dehydrogenase and 60S acidic ribosomal protein P2, which
ribosomal protein S3, T-complex protein 1, eta subunit and T-complex were less abundant in the secretome of the DL strain, have moonlighting
protein 1, theta subunit, presented higher abundance in DL secretome functions as protease and iron-binding protein, respectively (Table 2 and
than LCL one. Interestingly, an uncharacterized protein with increased Supplementary Table 4). In addition, the elongation factor 1-alpha, and
abundance in the secretome of DL strain (protein A4HAX5) has an Alba 40S ribosomal protein S3, which were more abundant in the secretome
domain (Table 2 and Supplementary Table 3), which has been involved of the DL strain, have alternative functions as cytoskeleton structure

7
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Table 1
Proteins identified in the secretion of L. (V.) braziliensis and classified in pro­
cesses that are relevant for parasite-host interaction.
Access number Name Number of
peptides

Stress response and protein


folding
A0A088RUF5 Glucose-regulated protein 78, 7
A4HII0 Heat shock 70-related protein 1, 12
mitochondrial
A4H9P0 Heat shock protein 10
A4HL70 Heat shock protein 83–1 18
A0A088RU16 Heat shock protein DNAJ, 5
A4HGY1 Heat-shock protein hsp70 21
A4HKX2 T-complex protein 1 subunit alpha 6
E9AIQ3 T-complex protein 1 subunit delta 5
E9AWE0 T-complex protein 1 subunit gamma 7
Fig. 4. Prediction of the secretion pathways for the proteins identified in the A4HN57 T-complex protein 1, eta subunit 4
L. (V.) braziliensis secretome. The figure shows the percentage of proteins with A4HQL2 T-complex protein 1, theta subunit 4
prediction for secretion via classical pathway (Signal P), alternative pathways A4H879 Tryparedoxin peroxidase 6
A4HKH1 Superoxide dismutase 3
(Secretome P), alternative-exosomal pathway (SecretomaP-exo), exosomal
E9JUH4 Trypanothione reductase 8
pathway (Exo), or without prediction.
A4HHC7 Tryparedoxin 4
A4HFW1 Trypanothione synthetase 1
regulator and DNA repair, respectively (Table 2 and Supplementary A4HCL7 Peroxidoxin 7
A0A088RZE8 Thiol-dependent reductase 1 5
Table 4). Interestingly, in L. donovani was described other alternative
Proteins involved in protein
function for the elongation factor 1-alpha as a peptidase [46], thus, for degradation
that protein, at least three different functions have been described. A0A088RI51 20S proteasome beta 6 subunit, 2
A4H6I8 Aminopeptidase 11
A4HMQ9 Aminopeptidase P 2
3.4. Functional interaction network of differentially abundant proteins in
A4HFH6 Calpain-like cysteine peptidase 25
the secretome of L. (V.) braziliensis strains Q4QCS4 Calpain-like cysteine peptidase, Clan 1
CA, family C2
To explore the biological processes of differentially abundant pro­ A4H716 Carboxypeptidase 11
teins in the secretome of DL and LCL strains, we also performed a A0A088RIB2 Dipeptidyl-peptidase III, 17
A4H638 GP63, leishmanolysin 15
functional interaction analysis. Our dataset of differential proteins was A4H5Q8 Oligopeptidase b 8
used to identify the orthologous proteins in Homo sapiens, and these A4HJV3 Phosphoglycan beta 1,3 6
were used to build a functional interaction analysis using the IIS- galactosyltransferase 5
interactome system web-accessible software. From the 26 differentially A4HQJ7 Prolyl oligopeptidase 3
A4HMA7 Proteasome activator protein pa26 3
abundant proteins, 15 presented orthologous in humans (Table 2), and
A4H9T4 Proteasome alpha 7 subunit 5
they were used for the interaction network (Fig. 5). The secretion of the A4HNP0 Proteasome beta 2 subunit 3
disseminated strain displayed two defined groups of proteins with A4HC34 Proteasome subunit alpha type 5
increased abundance: cellular protein metabolic processes (orthologs A4HP29 Proteasome subunit beta type 5
HSPA5, CCT8, CCT7) (Fig. 5), and gene expression (orthologs RPS3 and A4HF12 Thimet oligopeptidase 7
Proteins associated with
EEF1A1). Proteins with increased abundance (orthologs GOT1 and GSP) carbohydrate metabolism
and proteins with decreased abundance (orthologs MTAP, NME1, A4HQI8 2,3-bisphosphoglycerate-independent 6
AHCY, DLD) were grouped in the small molecule metabolic process cluster phosphoglycerate mutase
(Fig. 5). The remaining proteins were clustered in different functional A4H7T6 Enolase 9
A4HNY6 Fructose-bisphosphate aldolase 3
groups such as innate immune response, proteolysis, viral process, and
A4HMU1 Galactokinase-like protein 1
protein folding (Fig. 5). A4H643 Glycerol-3-phosphate dehydrogenase 7
Some proteins with increased abundance in the DL strain have direct [NAD(+)]
interactions between them: RPS3-YWHAE, RPS3-HSPA5 and CCT7- A4HFV1 Glycosomal phosphoenolpyruvate 2
CCT8 (Fig. 5). The first interaction corresponds to a functional associ­ carboxykinase
A7UFI6 Malate dehydrogenase 11
ation between gene expression (RPS3) and viral process (YWHAE); the A4HCQ1 NADP-dependent alcohol 2
second corresponds to the interaction between gene expression and pro­ dehydrogenase
tein metabolic processes (HSPA5); and the third one corresponds to an A4HBH9 Phosphoglycerate kinase 4
interaction between proteins involved in protein metabolic processes A0A088S299 Phosphomannomutase, 6
A4HM36 Pyruvate kinase 6
(CCT7 and CCT8). Interestingly, the proteins with increased abundance
A4H6M6 Pyruvate phosphate dikinase 2
showed a higher number of direct interactions with other proteins than A4H8J8 Transaldolase 4
those with reduced abundance. HSPA5 presented a total of 154 in­ A0A088SAQ9 Triosephosphate isomerase 6
teractions followed by EEF1A1 with 138, RPS3 with 121, and YWHAE A4HLU3 Udp-glc 4′ -epimerase 5
with 100 (Table 2). Amino acid metabolism
A4HQG9 Histidine secretory acid phosphatase 6
Most of the proteins with reduced abundance in the secretome of the A4H9Q9 Nonspecific nucleoside hydrolase 3
DL strain were not assigned to functional clusters but displayed in­ A0A088RTT3 Nucleoside 2-deoxyribosyltransferase, 3
teractions with different kinds of proteins (Fig. 5). We found, for A4HKT7 Nucleoside diphosphate kinase 8
example, that CALM1 (ortholog of calmodulin) contains the highest A4H4C5 S-methyl-5′ -thioadenosine 3
phosphorylase
number of interactions with other proteins (84 interactions), including
Nucleotide metabolism
proteins involved with regulation of gene expression and innate immune A4HPQ8 Adenosylhomocysteinase 16
response. In addition, three direct interactions were observed between (continued on next page)
increased (I) and decreased proteins(D): CALM1 (D)-YWHAE (I), DLD

8
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Table 1 (continued ) LCL strain. Conversely, the tyrosine aminotransferase (tat) and Glucose-
Access number Name Number of regulated protein 78 (Lpmp_281290) genes showed similar mRNA levels
peptides between the strains, whereas its protein abundance was significantly
A4HMB3 Aspartate aminotransferase 10
increased in the DL strain in comparison with the LCL strain (Fig. 6).
A0A088S0Y7 Cystathione gamma lyase, 5 Finally, we compared the gene expression of the uncharacterized protein
A4H3J2 Delta-1-pyrroline-5-carboxylate 4 (Lbrm_26_2610) that exhibited similar protein abundances between the
dehydrogenase DL and LCL strains in our dataset and, accordingly, the mRNA levels
Proteins involved in protein
were similar between the strains (Fig. 6).
translation and synthesis
A4H746 40S ribosomal protein S12 7
Q4Q9A5 40S ribosomal protein S16 2 4. Discussion
A4H3R3 40S ribosomal protein S19 protein 2
A4H9W3 40S ribosomal protein S2 2
Leishmania braziliensis is often associated with large clinical plas­
A4HLE6 40S ribosomal protein S3 4
A4HM77 40S ribosomal protein S3a 3
ticity, but the role of the parasite in such pleomorphism is not clear.
Q4QG31 40S ribosomal protein S4 3 Although the clinical outcome is a multifactorial phenomenon that in­
A4HMM1 40S ribosomal protein S6 1 volves several aspects of the host such as immune status, nutritional
A4H519 40S ribosomal protein S9 3 status, and genetic background, intrinsic aspects of the parasite may also
A4HQ28 40S ribosomal protein SA 2
modulate the pathogenic process and might influence the clinical form
E9AI48 60S acidic ribosomal protein 1
O44010 60S acidic ribosomal protein P2 2 of the disease [20,21,47]. However, it is not yet clear how and why some
A4H3Y8 60S ribosomal protein L10 3 strains of L. braziliensis spread metastatically while others do not. Here
A0A088SL38 60S ribosomal protein L10a, 3 we hypothesized that different strains modulate the activation of mac­
A4HDT2 60S ribosomal protein L12 2
rophages differentially and that such modulation would be one of the
A4HHP5 60S ribosomal protein L13 1
A4HAJ9 60S ribosomal protein L13a 1
first mechanisms contributing to the disparity in the clinical outcome.
A4H745 60S ribosomal protein L18 2 The release of molecules into the extracellular environment by par­
A4HK69 60S ribosomal protein L18a 1 asites of the Leishmania genus has been identified as one of the mecha­
A4HML2 60S Ribosomal protein L36 1 nisms used by this protozoan to evade or subvert the host’s immune
A4HMK9 60S ribosomal protein L5 3
response [27]. We observed essential changes in the parasite infection of
A4H873 60S ribosomal protein L6 3
A4H500 60S ribosomal protein L7a 1 macrophages pre-incubated with the secretome of each L. braziliensis
A4HFQ8 Arginyl-tRNA synthetase 5 strain. The pre-stimulation of peritoneal macrophages with secretome
A4H8V4 Elongation factor 1-alpha 6 significantly increases the percentage of infection, numbers of amasti­
Q2HZY7 Elongation factor 2 18
gotes per cell, and infection index over time. Those results suggested that
A4H5S5 Elongation factor-1 gamma 5
A4HCN4 Endoribonuclease L-PSP (Pb5) 4
the secretomes have proparasitic features that make macrophages more
A4HE05 Eukaryotic initiation factor 5a 3 susceptible to infection and favor the parasite proliferation in the early
A0A088S3S2 Eukaryotic translation initiation factor 2 stages of infection. Our data are in good agreement with previous studies
3 subunit 8, that showed that the treatment of C57BL/6 mice with L. donovani exo­
A4HGT5 Eukaryotic translation initiation factor 1
somes before to be challenged with the parasite exacerbated infection
3 subunit E
A4HNN3 Eukaryotic translation initiation factor 1 [48]. Also, co-inoculation of mice with L. major exosomes plus parasites
3 subunit L resulted in exacerbated lesions [43].
A4HNU9 Eukaryotic translation initiation factor 1 We also observed that the pre-treatment with the secretome of DL
6 strain resulted in a higher infection index than the pre-treatment with
Q25225 Probable eukaryotic initiation factor 11
4A
LCL secretome in all evaluated times, which suggested that the
A4HAJ7 Translation elongation factor 1-beta 6 biochemical composition of those secretomes might be different be­
tween the strains. The significant differences in the levels of proin­
flammatory cytokines secreted by macrophages pretreated and infected
(D)-DPP3 (I) and MTAP (D)-GOT1 (I). These direct interactions associate with each one of the strains reinforce such idea. We found that IL-6,
innate immune response with the cluster viral process (CALM1-YWHAE), TNF-α, and CCL2 levels were higher in macrophages infected with the
small molecule metabolic process with proteolysis (DLD-DPP3) and proteins LCL strain than in those infected with the disseminated one. This result
grouped in the category of metabolic process of small molecules (MTAP- supports the hypothesis that there is a strain-specific crosstalk between
GOT) (Fig. 5). each L.braziliensis strain and macrophages and, therefore, the composi­
Finally, the 894 direct interactions presented by the 15 differentially tion of the secretomes may also be distinct. Remarkably, IL-6 and TNF-α
abundant proteins (Fig. 5), indicate that there are a large number of concentration were two-fold higher in macrophages pre-treated with the
processes modulated by the secreted proteins and that different pro­ secretome of LCL strain concerning the same strain without pre-
cesses are affected by the proteins secreted by each strain. It is worth treatment, indicating that not only the direct parasite contact, but also
noting that proteins with increased abundance in the secretome of the the L. braziliensis secretome per se, is able to elicit macrophage responses.
DL strain showed a higher number of interactions when compared to Interestingly, LCL parasites, but not their secretome, were able to elicit
proteins increased in the LCL strain. IFN-γ responses, suggesting that direct contact between LCL parasites
and macrophages are necessary to IFN-γ production and reinforcing the
3.5. Gene expression of Lbrm_18_1400, lbrm_05_0800, and lip2 idea that LCL parasites activate responses in macrophages that
correlates with protein abundance contribute for the proper control of parasites. In contrast, neither DL
secretome nor parasites were able to elicit such responses.
To gain insights about the transcript levels of some of the differen­ Our results show that LCL secretome and/or parasites are able to
tially abundant proteins, we analyzed their gene expression by qPCR. In elicit the production of molecules involved in cell activation and
agreement with the relative quantification obtained by proteomics, the recruitment of monocyte and macrophages (IL-6, CCL2 and TNF-α) at
DL strain exhibited significant lower mRNA levels of the heat shock early times of stimulation/infection, whereas DL secretome and/or
protein (Lbrm_18_1400), S-methyl-5′ -thioadenosine phosphorylase parasites do not. Such results suggest that (i) the components of the LCL
(Lbrm_05_0800), and 60S acidic ribosomal protein P2 (Lip2) with a secretome that induce such markers are absent or at low abundance in
reduction of 3.8, 2.6 and 2.2-fold, respectively, when compared to the the DL secretome, or (ii) there are components in the DL secretome that

9
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Table 2
Differentially abundant proteins in the secretome of L. (V.) braziliensis strain associated with disseminated leishmaniasis compared to the strain associated with
localized cutaneous leishmaniasis.
Protein ID Name Number of Fold Stouffer P Abundance Secretion Orthology with Homo Number of
quantified Change value (D/I) pathway sapiens interactions
peptides
Gene Homo
name sapiens
code

A4HKT7 Nucleoside diphosphate kinase (EC 3 − 2.5 0.003 D Alternative - NME1 P15531 20
2.7.4.6) Exo
O44010 60S acidic ribosomal protein P2 2 − 2.3 0.007 D
(P2B-protein)
A4H440 Surface antigen-like protein 2 − 1.8 0.006 D Classical
A4HKX6 Dihydrolipoyl dehydrogenase (EC 2 − 1.8 0.017 D DLD P09622 20
1.8.1.4)
A4H5R9 Calmodulin 5 − 1.8 0.000 D Exo CALM1 P62158 84
A4H9P0 Putative heat shock protein 2 − 1.8 0.016 D HSPA4L O95757 17
A4H4C5 S-methyl-5′ -thioadenosine 2 − 1.6 0.005 D Alternative - MTAP Q13126 16
phosphorylase (EC 2.4.2.28) Exo
(MTAPase)
A4H746 40S ribosomal protein S12 2 − 1.6 0.026 D Alternative -
Exo
A4HPQ8 Adenosylhomocysteinase (EC 3 − 1.6 0.003 D Exo AHCY P23526 34
3.3.1.1)
A4H638 Leishmanolysin (EC 3.4.24.36) 5 − 1.6 0.001 D Alternative
A4H643 Glycerol-3-phosphate 3 − 1.6 0.004 D Alternative -
dehydrogenase [NAD(+)] (EC Exo
1.1.1.8)
A4H537 Splicing factor ptsr1-like protein 2 − 1.5 0.008 D Alternative
A4HFU7 Uncharacterized protein 4 − 1.5 0.001 D Alternative
A4HMB3 Putative aspartate aminotransferase 2 1.9 0.008 I GOT1 P17174 11
(EC 2.6.1.1)
A4HLE6 Putative 40S ribosomal protein S3 2 1.9 0.008 I Exo RPS3 P23396 121
A4HN57 T-complex protein 1 subunit eta 2 1.9 0.009 I Exo CCT7 Q99832 75
(TCP-1-eta)
A0A088RUF5 Glucose-regulated protein 78, 2 1.9 0.013 I Classical- HSPA5 P11021 154
putative Exo
A4HQL2 Putative T-complex protein 1, theta 2 1.8 0.008 I CCT8 P50990 78
subunit
A4H6F2 Putative 14–3-3 protein 3 1.8 0.002 I Exo YWHAE P62258 100
A4H3T9 Uncharacterized protein 6 1.7 0.000 I Classical
A4HPA3 TAT protein (EC 2.6.1.5) 8 1.7 0.000 I
A4H7H9 Thioredoxin-like_fold domain- 2 1.6 0.007 I
containing protein
E9JUH4 Trypanothione reductase 3 1.6 0.004 I Exo GSR P00390 12
(Fragment)
A4H8V4 Elongation factor 1-alpha 2 1.5 0.008 I Alternative - EEF1A1 P68104 138
Exo
A0A088RIB2 Dipeptidyl peptidase 3 (EC 2 1.5 0.008 I Exo DPP3 Q9NY33 14
3.4.14.4) (Dipeptidyl
aminopeptidase III)
A4HAX5 Contig, possible fusion of 3 1.5 0.011 I Alternative
chromosomes 20 and 34

abrogate the proper induction of these cytokines and chemokine. treatment with pentavalent antimonial than in samples from those who
The induction of higher levels of those molecules by LCL secretome/ failed [57]. TNF-α is a proinflammatory cytokine associated with a
parasites suggests that early activation and recruitment of monocytes by strong Th1 response and produced mainly by activated macrophages.
this strain could promote efficient parasite elimination. In contrast, DL This molecule is also essential to control parasite proliferation and
secretome/parasites failed to elicit such responses. Consistently, it has dissemination by the recruitment of cells to the site of infection and
been shown that IL-6 is highly expressed in localized cutaneous lesions subsequent formation of the granuloma [20,58,59]. Dendritic cells
[49], is upregulated in macrophages exposed to L.mexicana promasti­ exposed to L. braziliensis secreted molecules upregulate activation
gotes [50], and individuals with low macrophage IL-6 levels have markers, produce IL-12 and TNF-α and are more efficient at presenting
increased risk of developing mucocutaneus form, which implies para­ antigen [60]. However, in L. braziliensis infections, excessively high
site’s dissemination [51]. CCL2 is central for regulating the recruitment levels of TNF-α, together with a decreased ability of IL-10 to down­
and activation of macrophages and monocytes, and other cells required regulate this cytokine, can lead to severe tissue damage such as that
for early control of Leishmania infection [52–54]. Self-healing LCL is observed in patients with MCL [61,62].
associated with high levels of CCL2 in the skin lesions, where probably Interestingly, IL-10, which is recognized by its immunosuppressive
chemoattracts monocyte and macrophage and stimulates microbicidal role, was produced by macrophages pre-treated and/or infected with the
mechanisms [55]. In addition, in mice infected with L. donovani, CCL2 LCL strain, in all times evaluated. Downregulation of proinflammatory
favors the Th1 response by induction of IL-12 secretion and inhibition of molecules mediated by IL-10 could favor a wound healing response
IL-10 production in infected macrophages [56]. Recently, it was following parasite killing in infections with LCL strain, dampening the
observed that the expression of CCL2 is significantly higher in pre- immunopathogenic activity of cell recruitment and TNF-α production.
treatment lesion samples from patients who responded well to Consistently, it has been shown that IL-10 is important for

10
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

Fig. 5. Functional interaction network among differentially abundant proteins identified in the secretome of L. (V.) braziliensis strains. Proteins with differential
abundance in the secretome were clustered according to the biological process enriched with p < 0.05. Red circles: proteins with decreased abundance in the DL
strain in comparison with the LCL strain. Green circles: proteins with increased abundance in the DL strain in comparison with the LCL strain. The network was built
using the IIS (Integrated interactome system) platform and viewed in Cytoscape software version 2.8.3. (For interpretation of the references to colour in this figure
legend, the reader is referred to the web version of this article.)

Fig. 6. Gene expression analysis of selected genes with differential protein abundance in the secretome of two L. (V.) braziliensis strains. The mRNA levels of
lbrm_18_1400, lbrm_05_0800, lip2, tat, lpmp_281290, and lbrm_26_2610, were measured by qPCR in promastigotes of DL or LCL L. (V.) braziliensis strains. The values are
expressed as normalized relationships between the expression of the target gene and a geometric mean of two constitutive genes. Statistically analysis between the
strains were calculated using the Student t-test, using the GraphPad Prism 5.0 program. (*) p < 0.05 and (***) p < 0.0007. The graphs represent mean ± standard
deviation of biological triplicate.

11
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

counterbalance the exaggerated inflammatory response associated with environment [77–79]. In the case of cells of the immune system,
pathology in infections with L. braziliensis [63]. extracellular HSPs act as a signaling molecule for stress conditions,
Together, the induction of higher and sustained levels of IL-6, TNF-α, alerting other cells, thus preventing the extension of damage and pro­
and CCL2 by LCL strain, associated with balanced levels of IL-10, can moting resolution [78,80]. In human cell lines, extracellular HSP70
favor an effector cell response and a proper regulation of Th1 response exerts immunomodulatory functions, activating macrophages and
that result in the effective resolution of the disease caused by this strain. inducing the production of TNF-α [80]. Given the immunomodulatory
In contrast, the DL strain is not able to elicit such response or has functions of HSPs, it is possible to suggest that the low abundance of
mechanisms to prevent this activation, supporting efficient infection, HSP-A4H9P0 in the DL secretome, which was also decreased at the level
proliferation, and persistence of the parasite, which are indispensable of transcripts, could prevent the activation of proinflammatory path­
requirements for metastatic dissemination. Hence, despite macrophages ways, inhibiting the production of TNF-α and other cytokines. This
are programmed to destroy intracellular pathogens, their activation can proposition is supported by the almost undetectable levels of proin­
be modulated by the infectious agent resulting in successful colonization flammatory cytokines in macrophages pre-treated with DL secretome
of the intracellular environment that would otherwise be hostile. and/or infected with this strain, compared to the LCL strain. In fact, both
Results from several studies have shown that Leishmania spp. use the LCL secretome as well as the LCL parasite, were able to stimulate
different mechanisms to evade elimination by macrophages high levels of TNF-α, IL-6, and CCL2 in murine macrophages, indicating
[22,23,26,27,46,64]. Particularly, the interference of host signaling an appropriate activation of the cells to control the infection.
pathways prevents effective macrophage activation and favors para­ A metallopeptidase GP63 was also decreased in the DL secretome
sites’ survival and proliferation [22,46,65,66]. In order to obtain in­ when compared to the LCL secretome. This result is intriguing since it
formation about the molecules that could be associated to such has been shown that a GP63 isoform secreted by L.major can be inter­
modulation, we used a proteomics approach to identify and compara­ nalized by macrophages, activating different tyrosine phosphatases
tively quantify proteins secreted by LCL and DL strains. (PTPs), such as Src homology 2 domain containing tyrosine
Our dataset shows 252 proteins that were reproducibly identified in phosphatase-1 (SHP-1), protein tyrosine phosphatase 1B (PTP1B) and
the secretome of L. braziliensis. Various of those proteins have been protein tyrosine phosphatase T cell phosphatase (TCPTP), which induce
systematically reported in the secretions of Leishmania spp. including inactivation of the macrophage [22] and, consequently, favors the
HSP70, acid phosphatase, elongation factor 1β, tryparedoxin peroxi­ establishment of the parasite. Thus, considering that GP63 belongs to an
dase, and Leishmania homologue of activated C kinase (LACK), which are extensive gene family composed of more than 30 members with
involved in protein folding, regulation of the cell cycle and resistance to different potential functions in L. braziliensis [81], it is possible to sug­
oxidative stress [23–25]. We also observed that ~75% proteins secreted gest that the GP63 isoform identified here might play a different role
by L. braziliensis are exported by alternative pathways, mainly involving than that described in L. major [22]. In line with this proposition, we
exosomes, which is in good agreement with previous reports that showed in previous studies that strains associated to different forms of
showed that exosomes are a main route for protein secretion in Leish­ ATL exhibit different profiles of secreted metallopeptidases [82,83].
mania spp. [25,43,50,64,67,68]. Remarkably, several proteins identified Furthermore, the low abundance of this GP63 in the DL secretome
in the secretome have potential transmembrane domains, suggesting suggests the existence of alternative mechanisms of macrophage deac­
membrane localization, but various of them were also predicted to be tivation in this strain, such as those mediated by the elongation factor 1-
secreted by exosomes and are recurrently reported in the extracellular alpha 1 (EEF1A) [46].
milieu. Many of these proteins could be released by membrane shedding, In fact, we detected an increased abundance of EEF1A in the secre­
which is an important mechanism for communication in trypanosoma­ tome of DL strain compared to the LCL one. EEF1A catalyzes the binding
tids [69,70]. Consistently, L. donovani and L. mexicana secretomes also of aminoacyl-tRNAs to the ribosome site A during the growth of the
present proteins that are potentially targeted to membranes [24,71] or peptide chain but also plays a non-canonical function of peptidase.
intracellular compartments such as the mitochondrion, reinforcing the Peptidases can cleave and activate macrophage phosphatases that
idea that Leishmania proteins can perform more than one function in the inhibit the signaling cascade responsible for activating NO production,
intra- and extra-cellular space, similarly to the observed for other or­ favoring parasite establishment. Such a non-canonical function of
ganisms [72,73]. Our dataset still presented various proteins for which EEF1A as peptidase was observed in the secretion of L. donovani [46,66].
no prediction of conventional or alternative secretion was found, indi­ EEF1A activates SHP-1, a tyrosine phosphatase that negatively modu­
cating that there are several proteins with unknown routes of secretion/ lates the JACK/STAT (Janus kinase/signal transducer and activator of
releasing or that there is a limitation of the prediction tools for working transcription) and MAPK (mitogen-activated protein kinase) pathways
with these protozoa. resulting in the deactivation of the macrophage and decreased NO
We observed that the abundance of several proteins was significantly production [46,66,84]. Furthermore, in our functional analysis network,
reduced in the secretome of the DL strain compared to the LCL. Among EEF1A presented high complexity concerning the number of direct in­
those proteins, the 60S acidic ribosomal protein P2, which is involved in teractions, suggesting that the increase in its abundance would modulate
protein synthesis, has a non-canonical function as iron-binding protein, many processes. Additionally, we observed that the dipeptidyl-peptidase
and is associated with intracellular iron uptake [72,74]. As iron is III (DPP3) was also more abundant in the DL secretome. According to the
essential for the Leishmania replication [75], the increased expression of interaction network (A0A088RIB2), this peptidase affects critical pro­
molecules involved in iron uptake should favor its proliferation inside cesses, including activation of the MAPK pathway (UBC), cell death
the cell. However, infected macrophages rapidly upregulate the (NDRG1), and regulation of the apoptotic process (TNFAIP8). Therefore,
expression of iron transporters, sequestering the available metal, and the secretome of DL strain is enriched in peptidases that might deacti­
controlling parasite proliferation [75]. In this context, the potential vate macrophage’ signaling pathways crucial for parasite elimination.
decrease in iron uptake by DL strain could be a “silent” way of invading The glucose-regulated protein 78 (GRP78) was also more abundant
host cells, as it would not alert macrophages for overexpressing iron in the DL secretome than the LCL strain. This protein has also been found
transporters [75]. In addition, it was observed that limited iron condi­ in the secretome of L. donovani [24] and is actively secreted by cancer
tions induce Leishmania differentiation [76], which leads us to consider cells, and supports metastatic dissemination [85–87]. It has been sug­
that the disseminated strain may also have its differentiation favored gested that this protein can modulate parasite’s differentiation process
inside the host cells. [88], which is a fundamental requirement for Leishmania’s survival.
A heat shock protein (HSP-A4H9P0) was also less abundant in the Thus, our results suggest that DL strain could have higher efficiency in
secretome of DL strain than LCL strain. It is currently recognized that differentiation and proliferation, mediated by GRP78, which favors
HSPs have functions other than chaperones in the extracellular rapid adaptation to the phagolysosomal environment. As seen in cancer

12
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

cells, the secretion of this protein to the extracellular milieu could also Desenvolvimento Científico e Tecnológico - CNPq (P⋅C. Universal grant
promote the metastatic dissemination of parasites; however, this aspect No. 423300/2018–0); FIOCRUZ (P.C., A.R.V., M.L.B, N.P., G.P. PROEP
needs to be studied. grant No. 476727/2010–3); Fundação de Amparo à Pesquisa do Estado
Greater efficiency in the proliferation of DL strain could also be de Rio de Janeiro - FAPERJ (P.C. JCNE E-26/201.545/2014 and JCNE E-
mediated by the increase in aspartate aminotransferase, GOT1, which 26/203.253/2017; A.R.V. TCT No. E-26/202.539/2016), and Coor­
regenerates methionine from methylthioadenosine [89]. The synthesis denação de Aperfeiçoamento de Pessoal de Nível Superior – CAPES, −
of polyamines is fundamental for rapid cell proliferation and is a process Finance Code 001 (P.C Process No. 88887.374332/2019–00). G.P. was a
highly exploited by parasites and cancer cells [89,90]. The increase of CAPES fellow of the Visitant Professor Program (Process No. 0344141).
this protein in the secretion of the disseminated strain could favor the P.C, and J.B.J. are CNPq Researchers fellows (P.C. Process No. 306393/
rapid multiplication of the parasite in the first stages of infection in the 2014–0 and 305796/2017–8; J.B.J Process No. 309732/2018–2).
host cell, ensuring the establishment and dissemination of the infection All authors have read and approved the final manuscript.
[89,90].
The DL strain might also have a greater resistance to oxidative stress, Data availability
mediated by the increased abundance of trypanothione reductase, and
thioredoxin-like fold domain containing protein. Consistently, disrup­ The mass spectrometry proteomics data have been deposited to the
tion of the trypanothione reductase gene decreases the Leishmania ProteomeXchange Consortium via the PRIDE
infectivity and its ability to survive within the macrophage [91,92].
Besides, the resistance to oxidative stress has been shown to be associ­ Acknowledgements
ated with metastasis in mucocutaneous leishmaniasis caused by L.
panamensis and L.guyanensis strains [93]. Remarkably, the enriched We thank all the staff of the proteomic facilities of Fiocruz-Paraná
group of proteins involved in response to oxidative stress was abundant (Plataforma Proteômica RPT02H – FIOCRUZ) for technical help with
only in the DL strain, suggesting that this process may be crucial for mass spectrometric measurements, and Dr. Rosane Temporal and all the
dissemination. The mass spectrometry proteomics data have been staff of CLIOC for quality management advising.
deposited to the ProteomeXchange Consortium via the PRIDE [94]
partner repository with the dataset identifier #440425. Conflict of interest

5. Conclusions The authors declare no conflict of interests.

Using an iTRAQ-based quantitative proteomics approach we char­ References


acterized and compared the abundance of proteins secreted by L. bra­
ziliensis strains associated with very different clinical manifestations, DL [1] S. Burza, S.L. Croft, M. Boelaert, Leishmaniasis, Lancet 392 (2018) 951–970.
and LCL. Our dataset provides new information on molecules potentially [2] H.W. Murray, J.D. Berman, C.R. Davies, N.G. Saravia, Advances in leishmaniasis,
Lancet 366 (2005) 1561–1577.
involved in the process of infection and pathogenicity of those strains. [3] J. Alvar, I.D. Velez, C. Bern, M. Herrero, P. Desjeux, J. Cano, J. Jannin, M. den
Our data indicate that higher efficiency in detoxification and differen­ Boer, Team WHOLC, Leishmaniasis worldwide and global estimates of its
tiation can favor the infection and proliferation of the DL strain. In line incidence, PloS one 7 (2012), e35671.
[4] PAHO, Pan American Health Organization. Leishmaniasis: Epidemiological Report
with that evidence, macrophages pre-treated with the secretome of the in the Americas, PAHO, Washington, D.C, 2019.
DL strain have a higher infection index than those pre-treated with the [5] de Oliveira Camera P, Junger J, do Espirito Santo Silva Pires F, Mattos M, Oliveira-
LCL secretome. Besides, a better efficiency in the “silencing” of the Neto MP, Fernandes O, Pirmez C. Haematogenous dissemination of Leishmania
(Viannia) braziliensis in human American tegumentary leishmaniasis. Trans. R. Soc.
macrophage, mediated by peptidases, would further favor the estab­ Trop. Med. Hyg. 2006;100:1112–7.
lishment of infection and persistence of the DL strain. This “silencing” [6] L. Jirmanus, M.J. Glesby, L.H. Guimaraes, E. Lago, M.E. Rosa, P.R. Machado, E.
can also be corroborated by the reduced inflammatory response elicited M. Carvalho, Epidemiological and clinical changes in American tegumentary
leishmaniasis in an area of Leishmania (Viannia) braziliensis transmission over a 20-
in macrophages pre-treated with the secretome of the DL strain, and by
year period. Am. J. Trop. Med. Hyg. 86 (2012) 426–433.
the DL parasites per se, in comparison with the levels observed for the [7] P.D. Marsden, Mucosal leishmaniasis (“espundia” Escomel, 1911), Trans. R. Soc.
macrophages pre-treated and infected with the LCL strain. Thus, the DL Trop. Med. Hyg. 80 (1986) 859–876.
strain has mechanisms to prevent macrophage activation, favoring [8] J.E. Martinez, Arias L. Alba, M.A. Escobar, N.G. Saravia, Haemoculture of
Leishmania (Viannia) braziliensis from two cases of mucosal leishmaniasis: re-
parasite proliferation, and persistence, which are indispensable re­ examination of haematogenous dissemination, Trans. R. Soc. Trop. Med. Hyg. 86
quirements for metastatic dissemination. We also identified in the (1992) 392–394.
secretome of each strain several molecules that potentially mediated this [9] E. Thomaidou, L. Horev, D. Jotkowitz, M. Zamir, A. Ingber, C.D. Enk, V. Molho-
Pessach, Lymphatic dissemination in cutaneous Leishmaniasis following local
activation/deactivation and pointed out some molecules that could treatment, Am. J. Trop. Med. Hyg. 93 (2015) 770–773.
potentially be associated with the metastatic process. Additional studies [10] I. Bennis, S. Thys, H. Filali, V. De Brouwere, H. Sahibi, M. Boelaert, Psychosocial
of the secretome of other L. braziliensis strains need to be done to validate impact of scars due to cutaneous leishmaniasis on high school students in
Errachidia province, Morocco. Infect. Dis. Poverty. 6 (2017) 46.
the association of these molecules with the metastatic phenotype. [11] M. Yanik, M.S. Gurel, Z. Simsek, M. Kati, The psychological impact of cutaneous
Supplementary data to this article can be found online at https://doi. leishmaniasis, Clin. Exp. Dermatol. 29 (2004) 464–467.
org/10.1016/j.jprot.2020.104077. [12] C. Cincura, C.M.F. de Lima, P.R.L. Machado, J. Oliveira-Filho, M.J. Glesby, M.
M. Lessa, E.M. Carvalho, Mucosal leishmaniasis: a retrospective study of 327 cases
from an endemic area of Leishmania (Viannia) braziliensis, Am. J. Trop. Med. Hyg.
Author contributions 97 (2017) 761–766.
[13] A.L. Garcia, R. Parrado, E. Rojas, R. Delgado, J.C. Dujardin, R. Reithinger,
Leishmaniases in Bolivia: comprehensive review and current status, Am. J. Trop.
Designed the research: P⋅C., J.B.J., G.P.; conducted the research: A.R.
Med. Hyg. 80 (2009) 704–711.
V., M.L.B., L.R.B., C.M.R., A.C.B., P.A.; provided essential reagents/ [14] Y. Hashiguchi, E.L. Gomez, H. Kato, L.R. Martini, L.N. Velez, H. Uezato, Diffuse and
materials/analysis tools: P.C., R.M.B., P.C.C.; analyzed the data: P.C., A. disseminated cutaneous leishmaniasis: clinical cases experienced in Ecuador and a
brief review, Trop. Med. Health. 44 (2016) 2.
R.V., J.B.J.; wrote the manuscript: P.C., M.L.B. A.R.V. All authors have
[15] P.R. Machado, M.E. Rosa, D. Costa, M. Mignac, J.S. Silva, A. Schriefer, M.
read and approved the final manuscript. M. Teixeira, O. Bacellar, E.M. Carvalho, Reappraisal of the immunopathogenesis of
disseminated leishmaniasis: in situ and systemic immune response, Trans. R. Soc.
Funding Trop. Med. Hyg. 105 (2011) 438–444.
[16] M. Solomon, N. Sahar, F. Pavlotzky, A. Barzilai, C.L. Jaffe, A. Nasereddin,
E. Schwartz, Mucosal Leishmaniasis in travelers with Leishmania braziliensis
This work was supported by the Conselho Nacional de complex returning to Israel, Emerg. Infect. Dis. 25 (2019) 642–648.

13
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

[17] M.L. Turetz, P.R. Machado, A.I. Ko, F. Alves, A. Bittencourt, R.P. Almeida, [42] E. Willems, L. Leyns, J. Vandesompele, Standardization of real-time PCR gene
N. Mobashery, W.D. Johnson Jr., E.M. Carvalho, Disseminated leishmaniasis: a expression data from independent biological replicates, Anal. Biochem. 379 (2008)
new and emerging form of leishmaniasis observed in northeastern Brazil, J. Infect. 127–129.
Dis. 186 (2002) 1829–1834. [43] V.D. Atayde, H. Aslan, S. Townsend, K. Hassani, S. Kamhawi, M. Olivier, Exosome
[18] I.D. Velez, A. Jimenez, D. Vasquez, S.M. Robledo, Disseminated cutaneous secretion by the parasitic protozoan Leishmania within the sand Fly Midgut, Cell
Leishmaniasis in Colombia: report of 27 cases, Case reports in dermatology 7 Rep. 13 (2015) 957–967.
(2015) 275–286. [44] A. Dupe, C. Dumas, B. Papadopoulou, An Alba-domain protein contributes to the
[19] K.J. Gollob, A.G. Viana, W.O. Dutra, Immunoregulation in human American stage-regulated stability of amastin transcripts in Leishmania, Mol. Microbiol. 91
leishmaniasis: balancing pathology and protection, Parasite Immunol. 36 (2014) (2014) 548–561.
367–376. [45] A. Dupe, C. Dumas, B. Papadopoulou, Differential subcellular localization of
[20] C. Souza-Lemos, S.N. De-Campos, A. Teva, S. Corte-Real, E.C. Fonseca, R. Porrozzi, Leishmania Alba-domain proteins throughout the parasite development, PLoS One
G. Grimaldi Jr., Dynamics of immune granuloma formation in a Leishmania 10 (2015), e0137243.
braziliensis-induced self-limiting cutaneous infection in the primate Macaca [46] D. Nandan, T. Yi, M. Lopez, C. Lai, N.E. Reiner, Leishmania EF-1alpha activates the
mulatta, J. Pathol. 216 (2008) 375–386. Src homology 2 domain containing tyrosine phosphatase SHP-1 leading to
[21] C. Souza-Lemos, S.N. De-Campos, A. Teva, R. Porrozzi, G. Grimaldi Jr., In situ macrophage deactivation, J. Biol. Chem. 277 (2002) 50190–50197.
characterization of the granulomatous immune response with time in nonhealing [47] J.E. Martinez, B.L. Travi, A.Z. Valencia, N.G. Saravia, Metastatic capability of
lesional skin of Leishmania braziliensis-infected rhesus macaques (Macaca mulatta), Leishmania (Viannia) panamensis and Leishmania (Viannia) guyanensis in golden
Vet. Immunol. Immunopathol. 142 (2011) 147–155. hamsters, J. Parasitol. 77 (1991) 762–768.
[22] M.A. Gomez, I. Contreras, M. Halle, M.L. Tremblay, R.W. McMaster, M. Olivier, [48] J.M. Silverman, J. Clos, E. Horakova, A.Y. Wang, M. Wiesgigl, I. Kelly, M.A. Lynn,
Leishmania GP63 alters host signaling through cleavage-activated protein tyrosine W.R. McMaster, L.J. Foster, M.K. Levings, N.E. Reiner, Leishmania exosomes
phosphatases, Sci. Signal. 2 (2009) ra58. modulate innate and adaptive immune responses through effects on monocytes and
[23] B. Perez-Cabezas, N. Santarem, P. Cecilio, C. Silva, R. Silvestre, J. AMC, A. Cordeiro dendritic cells, J. Immunol. 185 (2010) 5011–5022.
da Silva, More than just exosomes: distinct Leishmania infantum extracellular [49] G. Caceres-Dittmar, F.J. Tapia, M.A. Sanchez, M. Yamamura, K. Uyemura, R.
products potentiate the establishment of infection, J. Extracell. Vesicles. 8 (2019) L. Modlin, B.R. Bloom, J. Convit, Determination of the cytokine profile in American
1541708. cutaneous leishmaniasis using the polymerase chain reaction, Clin. Exp. Immunol.
[24] J.M. Silverman, S.K. Chan, D.P. Robinson, D.M. Dwyer, D. Nandan, L.J. Foster, N. 91 (1993) 500–505.
E. Reiner, Proteomic analysis of the secretome of Leishmania donovani, Genome [50] K. Hassani, M. Olivier, Immunomodulatory impact of Leishmania-induced
Biol. 9 (2008) R35. macrophage exosomes: a comparative proteomic and functional analysis, PLoS
[25] P. Cuervo, J.B. De Jesus, L. Saboia-Vahia, L. Mendonca-Lima, G.B. Domont, Negl. Trop. Dis. 7 (2013), e2185.
E. Cupolillo, Proteomic characterization of the released/secreted proteins of [51] L. Castellucci, E. Menezes, J. Oliveira, A. Magalhaes, L.H. Guimaraes, M. Lessa,
Leishmania (Viannia) braziliensis promastigotes, J. Proteomics. 73 (2009) 79–92. S. Ribeiro, J. Reale, E.F. Noronha, M.E. Wilson, P. Duggal, T.H. Beaty, S. Jeronimo,
[26] N. Moradin, A. Descoteaux, Leishmania promastigotes: building a safe niche within S.E. Jamieson, A. Bales, J.M. Blackwell, A.R. de Jesus, E.M. Carvalho, IL6–174 G/C
macrophages, Front. Cell. Infect. Microbiol. 2 (2012) 121. promoter polymorphism influences susceptibility to mucosal but not localized
[27] U. Lambertz, J.M. Silverman, D. Nandan, W.R. McMaster, J. Clos, L.J. Foster, N. cutaneous leishmaniasis in Brazil, J. Infect. Dis. 194 (2006) 519–527.
E. Reiner, Secreted virulence factors and immune evasion in visceral leishmaniasis, [52] P. Allavena, G. Bianchi, D. Zhou, J. van Damme, P. Jilek, S. Sozzani, A. Mantovani,
J. Leukoc. Biol. 91 (2012) 887–899. Induction of natural killer cell migration by monocyte chemotactic protein-1, − 2
[28] M. Losada-Barragan, A. Umana-Perez, A. Rodriguez-Vega, S. Cuervo-Escobar, and − 3, Eur. J. Immunol. 24 (1994) 3233–3236.
R. Azevedo, F.N. Morgado, Carvalho V. de Frias, P. Aquino, P.C. Carvalho, [53] S. Oghumu, C.M. Lezama-Davila, A.P. Isaac-Marquez, A.R. Satoskar, Role of
R. Porrozzi, M. Sanchez-Gomez, G. Padron, P. Cuervo, Proteomic profiling of chemokines in regulation of immunity against leishmaniasis, Exp. Parasitol. 126
splenic interstitial fluid of malnourished mice infected with Leishmania infantum (2010) 389–396.
reveals defects on cell proliferation and pro-inflammatory response, J. Proteomics. [54] U. Ritter, H. Moll, Monocyte chemotactic protein-1 stimulates the killing of
208 (2019) 103492. Leishmania major by human monocytes, acts synergistically with IFN-gamma and is
[29] C. UniProt, UniProt: a worldwide hub of protein knowledge, Nucleic Acids Res. 47 antagonized by IL-4, Eur. J. Immunol. 30 (2000) 3111–3120.
(2019) D506–D515. [55] U. Ritter, H. Moll, T. Laskay, E. Brocker, O. Velazco, I. Becker, R. Gillitzer,
[30] P.C. Carvalho, J.S. Fischer, T. Xu, D. Cociorva, T.S. Balbuena, R.H. Valente, Differential expression of chemokines in patients with localized and diffuse
J. Perales, J.R. Yates 3rd, V.C. Barbosa, Search engine processor: filtering and cutaneous American leishmaniasis, J. Infect. Dis. 173 (1996) 699–709.
organizing peptide spectrum matches, Proteomics 12 (2012) 944–949. [56] R. Dey, N. Majumder, S. Bhattacharyya Majumdar, S. Bhattacharjee, S. Banerjee,
[31] P.C. Carvalho, J.S. Fischer, E.I. Chen, G.B. Domont, M.G. Carvalho, W.M. Degrave, S. Roy, S. Majumdar, Induction of host protective Th1 immune response by
J.R. Yates 3rd, V.C. Barbosa, GO explorer: a gene-ontology tool to aid in the chemokines in Leishmania donovani-infected BALB/c mice, Scand. J. Immunol. 66
interpretation of shotgun proteomics data, Proteome Sci. 7 (2009) 6. (2007) 671–683.
[32] P.C. Carvalho, D.B. Lima, F.V. Leprevost, M.D. Santos, J.S. Fischer, P.F. Aquino, J. [57] A. Navas, O. Fernandez, C. Gallego-Marin, M.D.M. Castro, M. Rosales-Chilama,
J. Moresco, J.R. Yates 3rd, V.C. Barbosa, Integrated analysis of shotgun proteomic J. Murillo, A. Cossio, D. McMahon-Pratt, N.G. Saravia, M.A. Gomez, Profiles of
data with PatternLab for proteomics 4.0, Nat. Protoc. 11 (2016) 102–117. local and systemic inflammation in the outcome of treatment of human cutaneous
[33] M.F. Carazzolle, L.M. de Carvalho, H.H. Slepicka, R.O. Vidal, G.A. Pereira, Leishmaniasis caused by Leishmania (Viannia), Infect. Immun. 88 (2020) e00764.
J. Kobarg, G.V. Meirelles, IIS–integrated Interactome system: a web-based platform [58] H.W. Murray, A. Jungbluth, E. Ritter, C. Montelibano, M.W. Marino, Visceral
for the annotation, analysis and visualization of protein-metabolite-gene-drug leishmaniasis in mice devoid of tumor necrosis factor and response to treatment,
interactions by integrating a variety of data sources and tools, PLoS One 9 (2014), Infect. Immun. 68 (2000) 6289–6293.
e100385. [59] P. Wilhelm, U. Ritter, S. Labbow, N. Donhauser, M. Rollinghoff, C. Bogdan,
[34] R.D. Finn, J. Mistry, J. Tate, P. Coggill, A. Heger, J.E. Pollington, O.L. Gavin, H. Korner, Rapidly fatal leishmaniasis in resistant C57BL/6 mice lacking TNF,
P. Gunasekaran, G. Ceric, K. Forslund, L. Holm, E.L. Sonnhammer, S.R. Eddy, J. Immunol. 166 (2001) 4012–4019.
A. Bateman, The Pfam protein families database, Nucleic Acids Res. 38 (2010) [60] L.P. Carvalho, E.J. Pearce, P. Scott, Functional dichotomy of dendritic cells
D211–D222. following interaction with Leishmania braziliensis: infected cells produce high levels
[35] T.N. Petersen, S. Brunak, G. von Heijne, H. Nielsen, SignalP 4.0: discriminating of TNF-alpha, whereas bystander dendritic cells are activated to promote T cell
signal peptides from transmembrane regions, Nat. Methods 8 (2011) 785–786. responses, J. Immunol. 181 (2008) 6473–6480.
[36] J.D. Bendtsen, L.J. Jensen, N. Blom, G. Von Heijne, S. Brunak, Feature-based [61] O. Bacellar, H. Lessa, A. Schriefer, P. Machado, A. Ribeiro de Jesus, W.O. Dutra, K.
prediction of non-classical and leaderless protein secretion, Protein Eng. Des. Sel. J. Gollob, E.M. Carvalho, Up-regulation of Th1-type responses in mucosal
17 (2004) 349–356. leishmaniasis patients, Infect. Immun. 70 (2002) 6734–6740.
[37] J.J. Almagro Armenteros, M. Salvatore, O. Emanuelsson, O. Winther, G. von [62] A.M. Da-Cruz, M.P. de Oliveira, P.M. De Luca, S.C. Mendonca, S.G. Coutinho,
Heijne, A. Elofsson, H. Nielsen, Detecting sequence signals in targeting peptides Tumor necrosis factor-alpha in human american tegumentary leishmaniasis, Mem.
using deep learning, Life Sci. Alliance. 2 (2019). Inst. Oswaldo Cruz. 91 (1996) 225–229.
[38] A. Krogh, B. Larsson, G. von Heijne, E.L. Sonnhammer, Predicting transmembrane [63] D.L. Costa, T.M. Cardoso, A. Queiroz, C.M. Milanezi, O. Bacellar, E.M. Carvalho, J.
protein topology with a hidden Markov model: application to complete genomes, S. Silva, Tr-1-like CD4+CD25-CD127-/lowFOXP3- cells are the main source of
J. Mol. Biol. 305 (2001) 567–580. interleukin 10 in patients with cutaneous leishmaniasis due to Leishmania
[39] S. Keerthikumar, D. Chisanga, D. Ariyaratne, H. Al Saffar, S. Anand, K. Zhao, braziliensis, J. Infect. Dis. 211 (2015) 708–718.
M. Samuel, M. Pathan, M. Jois, N. Chilamkurti, L. Gangoda, S. Mathivanan, [64] J.M. Silverman, N.E. Reiner, Leishmania exosomes deliver preemptive strikes to
ExoCarta: a web-based compendium of Exosomal cargo, J. Mol. Biol. 428 (2016) create an environment permissive for early infection, Front. Cell. Infect. Microbiol.
688–692. 1 (2011) 26.
[40] M. Losada-Barragan, A. Umana-Perez, S. Cuervo-Escobar, L.R. Berbert, R. Porrozzi, [65] J. Blanchette, N. Racette, R. Faure, K.A. Siminovitch, M. Olivier, Leishmania-
F.N. Morgado, D.A. Mendes-da-Cruz, W. Savino, M. Sanchez-Gomez, P. Cuervo, induced increases in activation of macrophage SHP-1 tyrosine phosphatase are
Protein malnutrition promotes dysregulation of molecules involved in T cell associated with impaired IFN-gamma-triggered JAK2 activation, Eur. J. Immunol.
migration in the thymus of mice infected with Leishmania infantum, Sci. Rep. 7 29 (1999) 3737–3744.
(2017) 45991. [66] D. Nandan, R. Lo, N.E. Reiner, Activation of phosphotyrosine phosphatase activity
[41] S.A. Bustin, J.F. Beaulieu, J. Huggett, R. Jaggi, F.S. Kibenge, P.A. Olsvik, L. attenuates mitogen-activated protein kinase signaling and inhibits c-FOS and nitric
C. Penning, S. Toegel, MIQE precis: practical implementation of minimum standard oxide synthase expression in macrophages infected with Leishmania donovani,
guidelines for fluorescence-based quantitative real-time PCR experiments, BMC Infect. Immun. 67 (1999) 4055–4063.
Mol. Biol. 11 (2010) 74.

14
A. Rodríguez-Vega et al. Journal of Proteomics 232 (2021) 104077

[67] J.M. Silverman, J. Clos, C.C. De’oliveira, O. Shirvani, Y. Fang, C. Wang, L.J. Foster, extracellular secretions of Leishmania (Viannia) braziliensis strains, Parasitology
N.E. Reiner, An exosome-based secretion pathway is responsible for protein export 132 (2006) 177–185.
from Leishmania and communication with macrophages, J. Cell Sci. 123 (2010) [83] P. Cuervo, A.L. Santos, C.R. Alves, G.C. Menezes, B.A. Silva, C. Britto,
842–852. O. Fernandes, E. Cupolillo, J. Batista De Jesus, Cellular localization and expression
[68] N. Santarem, G. Racine, R. Silvestre, A. Cordeiro-da-Silva, M. Ouellette, of gp63 homologous metalloproteases in Leishmania (Viannia) braziliensis strains,
Exoproteome dynamics in Leishmania infantum, J. Proteomics. 84 (2013) 106–118. Acta Trop. 106 (2008) 143–148.
[69] G. Arango Duque, A. Jardim, E. Gagnon, M. Fukuda, A. Descoteaux, The host cell [84] G. Forget, D.J. Gregory, L.A. Whitcombe, M. Olivier, Role of host protein tyrosine
secretory pathway mediates the export of Leishmania virulence factors out of the phosphatase SHP-1 in Leishmania donovani-induced inhibition of nitric oxide
parasitophorous vacuole, PLoS Pathog. 15 (2019), e1007982. production, Infect. Immun. 74 (2006) 6272–6279.
[70] M.F. Goncalves, E.S. Umezawa, A.M. Katzin, W. de Souza, M.J. Alves, B. Zingales, [85] J. Kern, G. Untergasser, C. Zenzmaier, B. Sarg, G. Gastl, E. Gunsilius, M. Steurer,
W. Colli, Trypanosoma cruzi: shedding of surface antigens as membrane vesicles, GRP-78 secreted by tumor cells blocks the antiangiogenic activity of bortezomib,
Exp. Parasitol. 72 (1991) 43–53. Blood 114 (2009) 3960–3967.
[71] K. Hassani, E. Antoniak, A. Jardim, M. Olivier, Temperature-induced protein [86] R. Langer, M. Feith, J.R. Siewert, H.J. Wester, H. Hoefler, Expression and clinical
secretion by Leishmania mexicana modulates macrophage signalling and function, significance of glucose regulated proteins GRP78 (BiP) and GRP94 (GP96) in
PLoS One 6 (2011), e18724. human adenocarcinomas of the esophagus, BMC Cancer 8 (2008) 70.
[72] L. Franco-Serrano, S. Hernandez, A. Calvo, M.A. Severi, G. Ferragut, J. Perez-Pons, [87] G. Zhao, J. Kang, K. Jiao, G. Xu, L. Yang, S. Tang, H. Zhang, Y. Wang, Y. Nie, K. Wu,
J. Pinol, O. Pich, A. Mozo-Villarias, I. Amela, E. Querol, J. Cedano, D. Fan, H. Zhang, D. Zhang, High expression of GRP78 promotes invasion and
MultitaskProtDB-II: an update of a database of multitasking/moonlighting metastases in patients with Esophageal squamous cell carcinoma, Dig. Dis. Sci. 60
proteins, Nucleic Acids Res. 46 (2018) D645–D648. (2015) 2690–2699.
[73] F. Meissner, R.A. Scheltema, H.J. Mollenkopf, M. Mann, Direct proteomic [88] N. Biyani, R. Madhubala, Quantitative proteomic profiling of the promastigotes
quantification of the secretome of activated immune cells, Science 340 (2013) and the intracellular amastigotes of Leishmania donovani isolates identifies novel
475–478. proteins having a role in Leishmania differentiation and intracellular survival,
[74] T. Furukawa, T. Uchiumi, R. Tokunaga, S. Taketani, Ribosomal protein P2, a novel Biochim. Biophys. Acta 1824 (2012) 1342–1350.
iron-binding protein, Arch. Biochem. Biophys. 298 (1992) 182–186. [89] L.C. Berger, J. Wilson, P. Wood, B.J. Berger, Methionine regeneration and aspartate
[75] C. Huynh, N.W. Andrews, Iron acquisition within host cells and the pathogenicity aminotransferase in parasitic protozoa, J. Bacteriol. 183 (2001) 4421–4434.
of Leishmania, Cell. Microbiol. 10 (2008) 293–300. [90] L.J. Marton, A.E. Pegg, Polyamines as targets for therapeutic intervention, Annu.
[76] B. Mittra, N.W. Andrews, IRONy OF FATE: role of iron-mediated ROS in Leishmania Rev. Pharmacol. Toxicol. 35 (1995) 55–91.
differentiation, Trends Parasitol. 29 (2013) 489–496. [91] C. Dumas, M. Ouellette, J. Tovar, M.L. Cunningham, A.H. Fairlamb, S. Tamar,
[77] S.K. Calderwood, J. Gong, A. Murshid, Extracellular HSPs: the complicated roles of M. Olivier, B. Papadopoulou, Disruption of the trypanothione reductase gene of
extracellular HSPs in immunity, Front. Immunol. 7 (2016) 159. Leishmania decreases its ability to survive oxidative stress in macrophages, EMBO
[78] A. De Maio, D. Vazquez, Extracellular heat shock proteins: a new location, a new J. 16 (1997) 2590–2598.
function, Shock 40 (2013) 239–246. [92] S. Krieger, W. Schwarz, M.R. Ariyanayagam, A.H. Fairlamb, R.L. Krauth-Siegel,
[79] E.A. Taha, K. Ono, T. Eguchi, Roles of extracellular HSPs as biomarkers in immune C. Clayton, Trypanosomes lacking trypanothione reductase are avirulent and show
surveillance and immune evasion, Int. J. Mol. Sci. 20 (2019). increased sensitivity to oxidative stress, Mol. Microbiol. 35 (2000) 542–552.
[80] V.L. Vega, M. Rodriguez-Silva, T. Frey, M. Gehrmann, J.C. Diaz, C. Steinem, [93] N. Acestor, S. Masina, A. Ives, J. Walker, N.G. Saravia, N. Fasel, Resistance to
G. Multhoff, N. Arispe, A. De Maio, Hsp70 translocates into the plasma membrane oxidative stress is associated with metastasis in mucocutaneous leishmaniasis,
after stress and is released into the extracellular environment in a membrane- J. Infect. Dis. 194 (2006) 1160–1167.
associated form that activates macrophages, J. Immunol. 180 (2008) 4299–4307. [94] Y. Perez-Riverol, A. Csordas, J. Bai, M. Bernal-Llinares, S. Hewapathirana, D.
[81] C. Yao, Major surface protease of trypanosomatids: one size fits all? Infect. Immun. J. Kundu, A. Inuganti, J. Griss, G. Mayer, M. Eisenacher, E. Perez, J. Uszkoreit,
78 (2010) 22–31. J. Pfeuffer, T. Sachsenberg, S. Yilmaz, S. Tiwary, J. Cox, E. Audain, M. Walzer, A.
[82] P. Cuervo, L. Saboia-Vahia, F. Costa Silva-Filho, O. Fernandes, E. Cupolillo, J.B. De F. Jarnuczak, T. Ternent, A. Brazma, J.A. Vizcaino, The PRIDE database and related
Jesus, A zymographic study of metalloprotease activities in extracts and tools and resources in 2019: improving support for quantification data, Nucleic
Acids Res. 47 (2019) D442–D450.

15

You might also like