Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Experimental Parasitology 210 (2020) 107846

Contents lists available at ScienceDirect

Experimental Parasitology
journal homepage: www.elsevier.com/locate/yexpr

Characterization of serum cytokines and circulating microRNAs that are T


predicted to regulate inflammasome genes in cutaneous leishmaniasis
patients
Lucilla Silva Oliveira Mendonçaa, Jaqueline Marques Santosb, Carla Martins Kanetoc,
Luciana Debortoli de Carvalhoc, Jane Lima-Santosc, Danillo G. Augustoc,d,
Silvia Maria Santos Carvalhoc, Jamária Adriana Pinheiro Soares-Martinse, Izaltina Silva-Jardimc,∗
a
Department of Health Sciences, Universidade Estadual de Santa Cruz (UESC), Ilhéus, BA, Brazil
b
Institute of Biomedical Sciences, Universidade de São Paulo (USP), São Paulo, SP, Brazil
c
Department of Biological Sciences, Universidade Estadual de Santa Cruz (UESC), Ilhéus, BA, Brazil
d
Department of Genetics, Universidade Federal do Paraná (UFPR), Curitiba, PR, Brazil
e
Department of Liberal Arts and Sciences, Milwaukee Area Technical College (MATC), Milwaukee, WI, United States

ARTICLE INFO ABSTRACT

Keywords: Leishmaniasis is a neglected disease caused by an intracellular protozoan parasite of the genus Leishmania.
Cutaneous leishmaniasis Infection starts when this protozoan replicates in a phagolysosomal compartment in macrophages, after evading
Cytokines host immune responses. The balance of Th1 and Th2 immune responses is crucial in leishmaniasis because it will
Inflammasomes determine whether the infection will be under control or if clinical complications will occur. The inflammasome,
microRNAs
which is activated during Leishmania infection, involves the action of caspase-1 and release of the proin-
flammatory cytokines interleukin-1β and interleukin-18. Together, they contribute to the maintenance of an
inflammatory response and pyroptosis. Here, we evaluated the serum levels of cytokines and the expression of
circulating microRNAs related to inflammasome regulation in twenty-seven patients with cutaneous leishma-
niasis in comparison to nine healthy individuals, in the context of the inflammasome activation. Evaluation of
serum cytokines activation (IL-1β, IL-2, IL-4, IL-6, IL-10, and IL-17) was performed by flow cytometry using CBA
kits (cytometric beads array) while the expression of circulating microRNAs (miR-7, miR-133a, miR-146b, miR-
155, miR-223, miR-328, and miR-342) in plasma was measured by quantitative polymerase chain reaction. Our
results showed an increase of the expression of miR-7-5p (p < 10−5), miR-133a (p = 0.034), miR-146b
(p = 0.003), miR-223–3p (p = 10−5), and miR-328–3p (p = 0.002), and cytokine levels for IL-1β (p = 0.0005),
IL-6 (p = 0.001), and IL-17 (p = 0.001) in patients with cutaneous leishmaniasis compared to the controls.
These results suggest that microRNAs and cytokines can play an important role in regulating the human immune
responses to Leishmania infection. Our findings may contribute to the understanding of the mechanisms of the
gene regulation during the cutaneous leishmaniasis and to the identification of possible biomarkers of the in-
fection.

1. Introduction most endemic countries in the Americas, with 12,690 cases of cuta-
neous (CL) and mucosal leishmaniasis (ML) (SisLeish-OPAS/WHO,
Leishmaniasis is a tropical parasitic disease caused by different 2018). Disseminated cutaneous leishmaniasis (DL) accounts for ap-
species of the protozoan Leishmania, which are transmitted to humans proximately 2% of the reported cases of CL in Brazil and diffuse cuta-
through the bites of the sand fly vector Phlebotomus (WHO, 2018). In neous leishmaniasis (DCL) another rare manifestation of CL with 1 or 2
Brazil, American cutaneous leishmaniasis (ACL) is one of the major cases diagnosed per year (Brasil, 2017). The ACL can be found in all
concerns regarding dermatological diseases because of its severity and regions of Bahia state, in which 67.2% of cities have been affected and
extensive human deformities (Brasil, 2017). an average of 2362 new cases are notified annually. The coefficient of
According to the 2016 epidemiological report, Brazil is among the detection was found to be at 32.8% risk of transmission in the Bahia


Corresponding author.
E-mail address: isjcavalli@uesc.br (I. Silva-Jardim).

https://doi.org/10.1016/j.exppara.2020.107846
Received 21 December 2018; Received in revised form 8 August 2019; Accepted 24 January 2020
Available online 28 January 2020
0014-4894/ © 2020 Elsevier Inc. All rights reserved.
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

state (SINAN and SUVISA, 2016). levels could help in the diagnosis, prognosis, and treatment of leish-
Depending on the host immune response and the parasite's entry, maniasis.
tropism, and pathogenicity, the disease can have outcomes ranging
from cutaneous lesions at the bite site, mucosal lesions to visceraliza-
tion, a more severe condition known as visceral leishmaniasis or kala 2. Material and methods
azar (Herwaldt, 1999). The main species of Leishmania found in de-
veloped countries cause cutaneous manifestations, which may either 2.1. Ethical aspects
heal spontaneously or progress to an intense inflammatory state,
making the treatment difficult (Herwaldt, 1999). The ACL may be This study was submitted to evaluation, and it was approved by the
subdivided into four types: i) localized cutaneous leishmaniasis (LCL), Ethics Committee on Human Research at UESC (CAAE
which is characterized as an ulceration with raised edges; ii) mucosal 18815013.7.1001.5526). Samples were only collected after patients
cutaneous leishmaniasis (MCL), which is a progression of the cutaneous were informed about the scope of this study and upon signing the in-
form and it is characterized by the destruction of the mucosa tissues, formed consent form. The study was conducted according to the
especially in the upper respiratory tract, iii) the rare form DL, which is Helsinki Declaration and Resolution 510/2016 of the National Health
characterized by the appearance of multiple polymorphic skin lesions in Council of the Ministry of Health that regulates studies involving hu-
various body regions, and most of the cases documented in the Amer- mans in Brazil.
icas are caused by Leishmania (V.) braziliensis (Brasil, 2017) and iv)
another atypical form of CL, the DCL, known as anergic-DCL, which is a
very rare and severe form of the disease with infiltrative nodular lesions 2.2. Population and sample collection
caused by L. (L.) amazonensis (Barral et al., 1995; Brasil, 2017; David
and Craft, 2009). Our study had 27 patients with active skin lesions and confirmed
During the course of the infection, the host's innate immunity plays diagnosis for cutaneous leishmaniasis that were enrolled prior to
an important initial role in the immune response by recruiting cells treatment. These patients were selected at the Health Center CAE III in
such as neutrophils, macrophages, and dendritic cells. These cells in- Ilhéus-BA, and they were diagnosed with CL based on the clinical
duce the production of reactive oxygen species (ROS) and release of characteristics of the disease along with the Montenegro test. Often,
nitric oxide (NO), in addition to pro-inflammatory cytokines that help these patients had one or a few ulcerative cutaneous lesions with raised
to fight the pathogen (Kedzierski and Evans, 2014). and delimited borders located in the lower limbs. Patients diagnosed
Cells from the innate immune system detect microorganisms with MCL were not included in this study. The control group was
through their pattern recognition receptors (PPRs) which bind to pa- comprised of 9 healthy individuals, in which they were grouped ac-
thogen-associated molecular patterns (PAMP), leading to a more spe- cording to their age, gender, and place of residence. The exclusion
cific immune response (Gurung and Kanneganti, 2016). NOD-like re- criteria for the control group included absence of skin lesions or in-
ceptors (NLRs), a family of PPRs, were shown to have a crucial role in fectious diseases and absence of previous history of leishmaniasis. The
defending the host against intracellular pathogens. NLRs play a role in information about our study population is presented in Table 1.
forming inflammasome complexes (Martinon and Tschopp, 2005). The We collected 8 mL (ml) of peripheral venous blood from patients
term inflammasome was initially described to determine a high mole- with cutaneous leishmaniasis and control group using vacuplast va-
cular weight complex that activates caspase-1 and it is responsible for cuum collection tubes®. We used 4 ml-tubes without additives and
processing of the proinflammatory cytokines IL-1β and IL-18 (Martinon containing a clot activator to separate the serum, and 4 ml-tubes with
et al., 2002). Inflammasomes can generate efficient signals and re- EDTA as an additive to separate the plasma.
sponses against pathogens by triggering the release of cytokines, acti-
vation of other immune cells, and programmed cell death (Di Virgilio,
2013).
Table 1
Some molecules can participate in the regulation of immune re-
Characteristics of participants at inclusion.
sponses, such as microRNAs (miRNAs) (Sutterwala et al., 2014). Mi-
croRNAs are small endogenous RNAs of approximately 22 non-coding Groups
nucleotides, which act as post-transcriptional regulators, cleaving ma-
Variables Control Patients Significance (p value)
ture mRNAs after binding to their untranslated 3′-end (He and Hannon,
2004). It is known that gene expression regulation by miRNAs also N % N %
impacts the expression of genes involved in the immune response. Some
Sex
miRNAs may have their expression increased in certain cell types, such
Male 5 55.6 16 59.3 NS (> 0.99)
as neutrophils, macrophages, and lymphocytes, ultimately regulating Female 4 44.4 11 40.7
several biological processes such as development, differentiation, and Age (years)
regulation of the immune system (Miska, 2005; Xiao and Rajewsky, 18–25 2 22.2 5 18.5
2009). 26–40 3 33.3 7 25.9 NS (0.91)
41–50 1 11.1 2 7.41
miRNAs play a modulatory role in the assembly of the inflamma- 51–60 2 22.2 5 18.5
some complex (Sutterwala et al., 2014), but the regulation of miRNAs 61–70 1 11.1 7 25.9
in leishmaniasis is still unclear. Some studies indicate the presence of Place of residence
unregulated miRNAs in human cells and cutaneous lesions, which af- Rural area 3 33.3 16 59.3 NS (0.25)
Urban area 6 66.7 11 40.7
fects the innate immune response (Diotallevi et al., 2018; Hashemi
Relapse
et al., 2018; Lemaire et al., 2013; Nunes et al., 2018). Yes NA NA 10 37
Up to this date, there are no reports showing the participation of No NA NA 17 63
miRNAs in the modulation of inflammasomes in cutaneous leishma- Presence of skin lesions
niasis, and this complex might play an essential role in the immune Yes 0 0 27 100 NA

response and tissue damage during the infection. In addition, miRNAs No 9 100 0 0
related to inflammasomes could help in the understanding the role of
this protein complex during infection. Furthermore, miRNAs could be Note: N-number samples. NA-not applicable. NS-not significant (Chi-Square
potential biomarkers for this disease, and detection of their circulating test).

2
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

2.3. Quantification of cytokines 2.6. Evaluation of the levels of circulating microRNAs in patients and
controls
Quantification of the cytokines IL-1β, IL-2, IL-4, IL-6, IL-10, and IL-
17 was performed using the CBA Flex Set System kit (BD®) in patients' The following microRNAs were evaluated in our study: hsa-miR-328
serum samples according to the manufacturer's instructions. Samples (TM 00543), hsa-miR-223 (TM 002098), hsa-miR-7 (TM 000268), hsa-
were first diluted at 1:4 ratio and the following assay reagents were miR-146b (TM001097), hsa-miR-155 (TM 002287), and hsa-miR-133a
prepared accordingly: standard reagents to calibrate the curve, fluor- (TM 002246). hsa-miR-342–3p (TM 002260) was used to normalize
ochrome detection reagent, and mix of the 8 different types of beads. miRNA input. microRNA expression was evaluated using quantitative
Briefly, 50 μL of mixed capture beads were added to 50 μL of polymerase chain reaction (qPCR). qPCR reactions contained pre-
standard samples or serum samples and incubated in the dark for 1 h at viously synthetized cDNA samples as template, specific probes for each
room temperature. After that, 50 μL of mixed detection reagents were microRNA, and TaqMan® Universal PCR Master Mix, in a final volume
added to the samples, followed by incubation in the dark for 2 h. of 20 μL qPCR reactions were performed in duplicate under the fol-
Samples were then, washed, centrifuged, and resuspended in 300 μL of lowing conditions: 50 °C for 2 min, 95 °C for 10 min, 40 cycles of 95 °C
washing buffer. Finally, samples were read in a flow cytometer (BD for 15 s, and 60 °C for 60 s using ABI 7500 Real Time PCR System.
FACSArray), calibrated with set-up beads. Analysis of results was done Levels of targeted miRNAs were normalized based on the endogenous
using the FCAP Array software. hsa-miR-342–3p, which showed a high expression level, with a low
standard deviation and constant values in both groups, being a suitable
normalizer for our samples according to (Liang et al., 2007). Quanti-
2.4. RNA extraction and complementary DNA synthesis (cDNA) fications of miRNAs levels were calculated using the 2-ΔCT method.

Total RNA was extracted from each plasma sample using TRIzol®
2.7. Prediction of target genes for microRNAs
reagent method (Life technologies, USA Molecular Research Center,
Inc).
MicroRNAs found to be differentially expressed were further eval-
RNA extractions were performed using 200 μL of plasma and 800 μL
uated using computational prediction on online bioinformatics plat-
of TRIzol under constant homogenization with micropipette in a tube.
forms to identify possible microRNAs-target genes interactions.
Then, 200 μL of chloroform were added to tubes followed by incubation
Because there are several bioinformatic platform tools and some of
at 4 °C for 5 min. After that, tubes were centrifuged at 15000 RPM at
them have different parameters, which can result in different interac-
4 °C for 15 min. Supernatants were transferred to a new tube and
tions, we selected three online bioinformatics tools widely used in
subsequently, the RNA was precipitated by adding 800 μL isopropanol
predictive studies (Targetscan-targetscan.org; Mirdb-mirdb.org and
to the aqueous phase. Samples were then placed at −80 °C overnight
Mirtarbase-mirtarbase.nbc.nctu.edu.tw), aiming to reduce the chances
and centrifugation at 15000 RPM at 4 °C for 20 min. Supernatants were
of false-positive results when predicting possible miRNAs target genes.
discarded, and pellets were washed through centrifugation with 1 mL of
For each online bioinformatic tool, we used the names of the dif-
70% ethanol. RNA pellets were resuspended in 30 μL RNAase-free
ferentially expressed microRNAs to determine possible target genes of
water. Subsequently, quantification was performed using Nanodrop®
each one. To improve the quality of results when predicting target
ND-1000 (Thermo Scientific-USA). Only RNA samples with a 260/280
genes, data were filtered through the intersection of the results of the
ratio of ≥1.8 were included.
three platforms, in which common target genes were found.
Complementary DNA synthesis (cDNA) was performed from RNA
After possible target genes were selected by intersecting results,
samples extracted from patients' blood (500ƞg) using miR-specific pri-
they were plotted in Cytoscape (available at http://www.cytoscape.
mers and Taqman miRNA Reverse Transcription Kit (Applied
org/) to obtain a graphic design of the interaction between microRNAs
Biosystem), in a scaled down volume of 15 μL RT reaction, according to
and their possible target genes. By doing so, we were able to have a
the manufacturer's instructions. Tubes were homogenized and in-
better depiction of microRNAs-target genes interactions. Then, using
cubated in the ThermoCycler (Techne Prime, BibbyScientific - UK). The
Cytoscape and BINGO (http://psb.ugente.be/cbd/papers/BINGO), we
thermal cycling parameters of reverse transcription were 30 min at
characterized the cellular processes that could occur due to the ex-
16 °C, 30 min at 42 °C and 5 min at 85 °C x 30 cycles. Subsequently,
pression of these genes.
samples were diluted at 1:4 ratio and stored at −20 °C, and when
needed, they used as template in quantitative PCR reactions.
2.8. Data analysis

2.5. microRNAs and inflammasomes For statistical analysis of cytokine levels and relative expression of
microRNAs in patients and control group, we applied the non-para-
We did an extensive search in the literature about microRNAs as- metric Mann-Whitney test (for two independent samples) using
sociated with the modulation of inflammation as shown in Table 2. GraphPad Prism (7.0) software. Results were considered significant

Table 2
MicroRNAs associated with inflammasomes and immune response mechanisms found in the in literature.
miR Description Reference

miR-7 - miR-7 as inhibitor of NLRP3 and IL-1β production during neuroinflammation in Parkinson's disease. Zhou et al. (2016).
- Suppression of NLRP3 and caspase-1 in neural stem cells. Fann et al. (2017).
miR-133 Regulation of inflammasome activation via suppression of UCP2 in THP cells. Bandyopadhyay et al. (2013).
miR-146 High expression of miR-146 downregulate both the expression of pro-inflammatory cytokines such as IL-1β and IL-18 and the Bhatt et al. (2016).
activation of inflammasomes in diabetic nephropathy.
miR-223 - The expression of miR-223 is downregulated when NLRP3 inflammasome and IL-1β are produced. Haneklaus et al. (2012).
- The activation of inflammasomes is negatively regulated by miR-223. Franz Bauernfeind et al. (2012).
- miR-223 inhibits the production of caspase-1, NLRP3, and IL-1β during intracerebral hemorrhage in animal models. Yang et al. (2015b).
miR-328 miR-328 is differentially expressed in macrophages infected with Leishmania donovani, suggesting that it may play a role in Tiwari et al. (2017)
phagocytosis.

3
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

Fig. 1. Quantification of cytokines in the sera of patients with active lesions of cutaneous leishmaniasis (CT). A – F) Levels of serum cytokines IL-1β, IL-2, IL-4, IL-6,
IL-10, IL-17. (Graphs show the mean of cytokine production using the non-parametric Mann-Whitney t-Test).

when p-value was < 0.05. We used the R software for Spearman cor- convergence of three different prediction algorithms, our results
relation analysis tests (for the correlation matrix) and simple x-y plot. showed that these miRNAs possibly regulate several genes related to the
To determine the homogeneity in our group population, we per- immune response, such as those involved in the regulation of pro-
formed a Chi-square statistical test (p > 0.05), which showed that grammed cell death (DNAJB6, IRS2, DNAJC5, RBPJ, IGF1R, FOXO3,
both groups are similar. ECT2, MEF2C, FOXO1, and TGFB2), caspase activity (NLRP3, FOXL2,
F3, SENP1, and SNCA), and response to cytokine stimuli (IRAK1,
3. Results TRAF6, IL-6ST, MCL1, BCL2L1) (Fig. 3).
Altogether, our results showed an up-regulation of microRNAs as-
3.1. Levels of IL-1β, IL-6, and IL-17 were increased in the serum of patients sociated with inflammasomes in plasma of patients with cutaneous
with cutaneous leishmaniasis leishmaniasis when compared to healthy controls.

We quantified the levels of serum cytokines (IL-1β, IL-6, and IL-17) 3.3. Correlation between levels of miRNA and cytokines in the
in patients with cutaneous leishmaniasis (CL) and compared to healthy inflammasome activation
controls. Our results showed significant increase in the cytokines levels
for IL-1β (p = 0.0005), IL-6 (p = 0.001), and IL-17 (p = 0.001) in To understand the correlation between the levels of miRNAs and
patients (Fig. 1). Increased cytokines levels in patients with LC may cytokines found in our study in the context of inflammasomes, we did a
trigger a Th17 immune response and an increase in IL-1β levels suggests correlation matrix (Fig. 4a). We found an inversely proportional cor-
the activation of inflammasomes. relation between levels of IL-1β and the microRNAs miR-223 and miR-
7, whereas levels of miR-133a, miR-146b, and miR-328 presented po-
3.2. Quantification of microRNAs (miR-7-5p, miR-133a, miR-146b, miR- sitive values compared to IL-1β levels.
223, and miR-328–3p) in plasma of individuals with cutaneous Based on both the literature and computational prediction, our
leishmaniasis analyses indicated that miR-7, miR-223, and miR-133a played an im-
portant role in the inflammasome activation. The correlation between
Based on the literature, we found five miRNAs that possibly regulate the levels of these miRNAs and IL-1β in our patients’ samples showed
genes involved in inflammasomes activation. Our results showed in- that the higher the levels of IL-1 β, the lower the levels miR-7 and miR-
creased levels of miR-7-5p, miR-133a, miR-146b, miR-223, and miR- 223, whereas levels of miR-133a correlate positively cytokine levels, as
328–3p in plasma of patients with cutaneous leishmaniasis (p < 0.05; shown in Fig. 4b–d.
Fig. 2). The most significant differences were found for miR-7-5p
(p < 10−5) and miR-223 (p = 10−5), in which their relative expres- 4. Discussion
sions were 10,000 and 3.8-fold up in patients with cutaneous leish-
maniasis, respectively, whereas miRNAs miR133a, miR 328-3-P and Inflammation is a crucial factor in the development of lesions during
miR146b presented p-values equal to 0.034, 0.002, and 0.003, respec- Leishmania infection (Kaye and Scott, 2011). Assembly and activation of
tively. the inflammasome complex during Leishmania infection has already
To understand the role of these miRNAs in gene regulation, we used been demonstrated (Gurung et al., 2015; Lima-Junior et al., 2013). It
in silico prediction to search for their possible targets. After the has been suggested this activation is important for the healing process

4
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

Fig. 2. Serum levels of the microRNAs miR-7, miR-133a, miR-146b, miR-223, and miR-328-3p in patients with American cutaneous leishmaniasis (ACL)
and individuals from the control group using quantitative PCR. A–E: serum levels of miR-7-5p, miR-133a, miR-146b, miR-223, and miR-328–3p, respectively.
Data show means and standard deviations using the non-parametric Man-Whitney U Test.

by stimulating the synthesis of nitric oxide and fighting of parasite Bauernfeind et al., 2012; Gupta et al., 2017; Liu et al., 2015a). In addition,
(Lima-Junior et al., 2013). However, this is controversial as other stu- we identified possible target genes in the miR-146b, such as TRAF6 and
dies have shown that activated inflammasomes actually contribute to IRAK1, which are known to be involved in the immune response mediated
the severity of this disease (Gurung et al., 2015; Novais et al., 2017). by TLRs (toll like receptors) (Park et al., 2015). It was important to predict
Depending on the Leishmania species and the host cellular immune possible genes in the microRNAs miR-7 and miR-328 because they are
response, different cytokines and chemokines can be produced. In this involved in processes of cell death, phagocytosis, and autophagy (Tay
study, a significant difference was observed in serum cytokines levels (IL- et al., 2015; Wang et al., 2017).
1β, IL-6, and IL-17) from patients with CL compared to the control group, Most studies involving miR-146 focused on inflammatory roles, and
which indicates a similar immune response during leishmaniasis (Espir this microRNA is extensively described as one of the induced miRNAs in
et al., 2014; Gomes et al., 2014). From all cytokines, we also found greater response to cytokines and pathogenic products in macrophages and
levels for IL-1β, which suggests activation of inflammasomes. dendritic cells (Marques-Rocha et al., 2015; Park et al., 2015). This
IL-1β is an important mediator of innate and adaptive immune re- miRNA plays a role as an anti-inflammatory regulator in various types
sponses, and its association with inflammasomes activation has been of immune cells by suppressing NF-κβ signaling and stimulating a ne-
demonstrated. Exacerbation of IL-1β production and secretion may gative feedback. Increased levels of miR-146 lead to the silencing of
trigger the inflammatory process that makes difficult to heal the lesions IRAK1 and TRAF genes (Echavarria et al., 2015; Park et al., 2015).
in cutaneous leishmaniasis (Novais et al., 2017; Peniche et al., 2017). miR-328 may have a role in regulating innate immune cells, which
However, IL-1β may also be involved in the severity of other forms of can inhibit cell growth and promote apoptosis through PAK6 due to an
the disease, as in the case of the diffused form caused by Leishmania increase in caspases 3 and 9 levels (Liu et al., 2015b). When miR-328
mexicana (Fernández-Figueroa et al., 2012). expression is inhibited, phagocytosis increase and chances that patho-
IL-1β levels may be directly related to both the increase in IL-6 le- gens will survive are reduced (Tay et al., 2015). microRNAs can
vels and triggering of type Th17 immune response with IL-17 produc- downregulate the PTPRJ gene that encodes for the protein tyrosine
tion (Dinarello, 2011). IL-6 is a potent inflammatory cytokine that plays phosphatase (PTP); dysfunction of tyrosine phosphorylation is related
a role in acute immune response. Curing infections or tissue lesions, IL- to immune disorders (Paduano et al., 2012). In leishmaniasis, induction
6 mediates several physiological functions such as lymphocytes differ- of PTP expression by the parasite can lead to a negative regulation of
entiation and cell death mechanisms. IL-17 stimulates the secretion of host cell functions (Blanchette et al., 1999), in addition to the sup-
IL-6 and IL-8 in human fibroblasts and also participates in several mi- pression of matrix metalloproteinase-2 (MMP2) (Yang et al., 2015a).
crobial diseases and autoimmune diseases (Bettelli et al., 2007). Increased expression of MMP2 are associated with successful healing
Our data showed a differential expression of the microRNAs miR-7-5p, Leishmania lesions (Maretti-Mira et al., 2010).
miR-133a, miR-146b, miR-223, and miR 328–3p, which may be related to Studies performed in vivo and in vitro showed that miR-7-5p is a
pathogen-host interaction in cutaneous leishmaniasis. When predicting negative regulator of inflammasomes activation by significantly and
possible target genes, we identified several that were related to in- specifically decreasing the levels of the inflammasome protein NLRP3
flammasomes, such as NLRP3 in the miR-223, a protein involved in in- (Zhou et al., 2016). There are studies that demonstrated the partici-
flammasome assembly, and BCL2L1 and UCP2 in the miR-133a, an anti- or pation of miR-7-5p in the modulation of IL-1β gene, which suggests that
pro-apoptotic regulator involved in NLR receptor signaling and a protein this microRNA may play a key role in the induction of this cytokine (Liu
that negatively controls inflammasomes activities, respectively (F. et al., 2016).

5
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

(caption on next page)

6
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

Fig. 3. Computational prediction for miR-7, miR-133a, miR-146b, miR-223, and miR-328-3p. A–E: Representation of the interaction of possible target genes
with miRNAs. Genes were selected based on the intersection of results obtained from three different online bioinformatics platforms (TargetScan, miRDB e
miRTarBase) and then, distributed in the integration network using Cytoscape Computational Program. Yellow ellipse represents miRNAs. Blue rectangles represent
possible target genes predicted in general. Green rectangles represent possible predicted target genes involved in programmed cell death. Red rectangles represent
possible predicted genes involved in caspase activity. Pink rectangles represent possible predicted genes involved in response to cytokine stimuli. (For interpretation
of the references to colour in this figure legend, the reader is referred to the Web version of this article.)

miR-223 is described as a regulator of the inflammasome protein (Bandyopadhyay et al., 2013). Deficiency of UCP2 gene increases the
NLRP3, targeting the protein binding site. When expression levels of activation of the inflammasome protein NLRP3 by activating reactive
miR-223 are increased, inflammasome activation is compromised be- oxygen species (ROS). Expression of UCP2 gene was found to reduce
cause levels and accumulation of the protein NLRP3 are decreased. NLRP3 gene expression and IL-1β and IL-18 levels, but it did not affect
However, other studies suggest that miR-223 expression has an oppo- the expression of the inflammasome protein AIM2 (Moon et al., 2015).
site association with NLRP3 protein, and it might be a negative reg- In addition, miR-133a also targets MCL1 and BCLXL (BCL2L1 coding
ulator of inflammasome (F. Bauernfeind et al., 2012; Chen and Sun, gene) genes, which are inhibitors of apoptosis, caspase-1, and release of
2013; Yang et al., 2015b). Assays performed in vitro and in vivo have IL-1β (Liu et al., 2015a). Thus, these results suggest that miR-133a
shown that the inflammasome protein NLRP3 is activated in response to promotes inflammasomes activation.
Leishmania infection, and it is important to inhibit parasite's replication Inflammasomes activation is critical during host defense mechan-
in macrophages. isms (Osawa et al., 2011). Still, in leishmaniasis, the role of in-
miR-133a participates in silencing the UCP2 gene, which can reg- flammasomes in the host defense mechanism against Leishmania is not
ulate the activation of inflammasomes. In a study with THP1 cells, well understood. Recent studies using resistant mice showed that
where UCP2 gene was silenced, an increase in inflammasome activa- during L. amazonensis infection inflammasomes are important. How-
tion, caspase-1 level, and IL-1β secretion was observed when compared ever, during infections caused by L. major, L. mexicana, and L. donovani,
to cells that exhibited greater expression of this gene. These results inflammasomes activation might be inhibited by these parasites, con-
suggest that UCP2 plays a role as a negative regulator of inflamma- tributing to the establishment of a virulent infection (Lima-Junior et al.,
somes activation whereas miR-133a plays a role as a positive regulator 2013; Shio et al., 2015).

Fig. 4. Correlation between levels of cytokines and microRNAs. A – Correlation matrix between levels of all cytokines and miRNAs. Circles and corresponding
sizes represent the significance level. Colors represent the directionality of the correlation (blue - positive correlations; red - negative correlations) B – The correlation
between levels of IL-1β and miR-133a indicates a positive correlation. C- The correlation between levels of IL-1β and miR-7, indicates a negative correlation. D - IL-1β
and miR-223 levels indicate a negative correlation. (For interpretation of the references to colour in this figure legend, the reader is referred to the Web version of this
article.)

7
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

Pathogens evolved to modulate immune responses, such as the ac- Declaration of competing interest
tivation of inflammasomes by preventing its assembly (Lamkanfi and
Dixit, 2012). One of the proposed mechanisms that leads to inhibition The authors declare that they have no competing interests.
of the inflammasome protein NLRP3 involves the prostaglandins (PG)
signaling pathway. Specifically, PGE2 suppresses the activation of the Acknowledgments
inflammasome protein NLRP3 via induction of cyclic adenosine
monophosphate (cAMP), which binds to protein kinase A (PKA), re- Authors thank patients for participating in this study and SMS-
sulting in NLRP3 phosphorylation and inactivation of inflammasomes CAEIII employees in Ilhéus-BA for their support during collection of
(Mortimer et al., 2016). Leishmania donovani is able to induce PGE2 in samples.
macrophages by stimulating cyclooxygenase 2 (COX2), an enzyme in-
volved in inflammation and production of prostaglandins. The later can Appendix A. Supplementary data
act as immunosuppressants during infection (Matte et al., 2001).
Studies have shown induction of PGE2 by Leishmania species that Supplementary data to this article can be found online at https://
cause the cutaneous disease form, in which animals infected with doi.org/10.1016/j.exppara.2020.107846.
Leishmania amazonensis were treated with prostaglandin antagonists
and parasitemia decreased (Guimarães et al., 2006). Furthermore, it has References
been shown that patients with cutaneous leishmaniasis have higher
levels of plasma PGE2 compared to healthy individuals, suggesting that Bandyopadhyay, S., Lane, T., Venugopal, R., Parthasarathy, P.T., Cho, Y., Galam, L.,
prostaglandins are involved in the course of infection and they have an Lockey, R., Kolliputi, N., 2013. MicroRNA-133a-1 regulates inflammasome activation
through uncoupling protein-2. Biochem. Biophys. Res. Commun. 439, 407–412.
important role in the persistence of the pathogen and modulation of the https://doi.org/10.1016/j.bbrc.2013.08.056.
host immune response (França-Costa et al., 2015). Barral, A., Costa, J.M.L., Bittencourt, A.L., Barral‐Netto, M., Carvalho, E.M., 1995. Polar
Our results showed an increase in cytokines levels in the sera of and subpolar diffuse cutaneous leishmaniasis in Brazil: clinical and im-
munopathologic aspects. Int. J. Dermatol. 34, 474–479. https://doi.org/10.1111/j.
patients with American cutaneous leishmaniasis (ACL), especially for 1365-4362.1995.tb00613.x.
IL-1β, which is an important cytokine that can be produced and se- Bauernfeind, F., Rieger, A., Schildberg, F.A., Knolle, A., Schmid-burgk, J.L., Hornung, V.,
creted upon inflammasomes activation. Furthermore, increased IL-6 Schmid-burgk, J.L., Hornung, V., 2012. NLRP3 inflammasome activity is negatively
controlled by miR-223. https://doi.org/10.4049/jimmunol.1201516.
and IL-17 levels may be induced via NOD2 receptor by Leishmania
Bettelli, E., Oukka, M., Kuchroo, V.K., 2007. TH-17 cells in the circle of immunity and
species that cause the cutaneous form of the disease (Dos Santos et al., autoimmunity. Nat. Immunol. 8, 345–350. https://doi.org/10.1038/ni0407-345.
2017). The differential expression of miR-133a has been described as a Bhatt, K., Lanting, L.L., Jia, Y., Yadav, S., Reddy, M.A., Magilnick, N., Boldin, M.,
Natarajan, R., 2016. Anti-inflammatory role of MicroRNA-146a in the pathogenesis of
positive regulator of inflammasomes activation. However, other circu-
diabetic nephropathy. J. Am. Soc. Nephrol. 27, 2277–2288. https://doi.org/10.1681/
lating microRNAs, such as miR-7-5p and miR-223, were identified to ASN.2015010111.
negatively regulate inflammasomes. Blanchette, J., Racette, N., Faure, R., Siminovitch, K.A., Olivier, M., 1999. Leishmania-
In leishmaniasis, the host inflammatory response is important to con- induced increases in activation of macrophage SHP-1 tyrosine phosphatase are as-
sociated with impaired IFN-γ-triggered JAK2 activation. Eur. J. Immunol. 29,
trol the infection and parasite replication. Our innate immune system is 3737–3744. https://doi.org/10.1002/(SICI)1521-4141(199911)29:11<3737::AID-
able to fight pathogens by assembling complexes such as the inflamma- IMMU3737>3.0.CO;2-S.
some and it may be that certain miRNAs negatively regulate inflamma- Brasil, da saúde, M., 2017. Manual de vigilância da leishmaniose tegumentar, ministério.
In: Secretaria de Vigilância em Saúde. Departamento de Vigilância das Doenças
somes, which may contribute to pathogens’ immune evasion mechanisms, Transmissíveis, (Brasilia).
ultimately inactivating inflammasomes. Our study showed the role of Chen, S., Sun, B., 2013. Negative regulation of NLRP3 inflammasome signaling. Protein
microRNAs target genes in inflamasomes activation, such as miR-7 and Cell 4, 251–258. https://doi.org/10.1007/s13238-013-2128-8.
David, C., Craft, N., 2009. Cutaneous and mucocutaneous leishmaniasis. Dermatol. Ther.
miR-223, which may inhibit the complex and facilitate the spread of the 22, 491–502. https://doi.org/10.1111/j.1529-8019.2009.01272.x.
parasite. These miRNAs were detected at higher levels in patients with Di Virgilio, F., 2013. The therapeutic potential of modifying inflammasomes and NOD-
lower levels of IL-1 β, whereas patients with increased levels of this cy- like receptors. Pharmacol. Rev. 65, 872–905. https://doi.org/10.1124/pr.112.
006171.
tokine also had a proportional increase in miR-133a levels, which may Dinarello, C.A., 2011. Interleukin-1 in the pathogenesis and treatment of inflammatory
indicate that this microRNA is important in the activation of the in- diseases. Blood 117, 3720–3732. https://doi.org/10.1182/blood-2010-07-273417.
flamasome and may contribute to resolution of this disease. Diotallevi, A., Santi, M. De, Buffi, G., Ceccarelli, M., Vitale, F., Galluzzi, L., Magnani, M.,
2018. Leishmania infection induces MicroRNA hsa-miR-346 in human cell line-de-
In sum, our results showed that certain microRNAs are differentially
rived macrophages. Front. Microbiol. 9, 1–9. https://doi.org/10.3389/fmicb.2018.
expressed during cutaneous leishmaniasis, suggesting that they can be 01019.
used as possible biomarkers of the disease, especially because studies Dos Santos, J.C., Damen, M.S.M.A., Oosting, M., De Jong, D.J., Heinhuis, B., Gomes, R.S.,
involving miRNAs and leishmaniasis are still scarce. Our data also Araújo, C.S., Netea, M.G., Ribeiro-Dias, F., Joosten, L.A.B., 2017. The NOD2 receptor
is crucial for immune responses towards New World Leishmania species. Sci. Rep. 7,
showed an increase in pro-inflammatory cytokines levels (IL-1β, IL-6, 1–10. https://doi.org/10.1038/s41598-017-15412-7.
and IL-17), suggesting that these cytokines and microRNAs participate Echavarria, R., Mayaki, D., Neel, J.C., Harel, S., Sanchez, V., Hussain, S.N.A., 2015.
in the regulation of the host immune response during cutaneous leish- Angiopoietin-1 inhibits toll-like receptor 4 signalling in cultured endothelial cells:
role of miR-146b-5p. Cardiovasc. Res. 106, 465–477. https://doi.org/10.1093/cvr/
maniasis, and they might play a role in modulating the response to cvv120.
inflammasomes activation. Espir, T.T., Figueira, L.D.P., Naiff, M.D.F., Da Costa, A.G., Ramalho-Ortigão, M., Malheiro,
A., Franco, A.M.R., 2014. The role of inflammatory, anti-inflammatory, and reg-
ulatory cytokines in patients infected with cutaneous leishmaniasis in amazonas
Authors' contributions state, Brazil. J. Immunol. Res. 2014. https://doi.org/10.1155/2014/481750.
Fann, D.Y.-W., Lim, Y.-A., Cheng, Y.-L., Lok, K.-Z., Chunduri, P., Baik, S.-H., Drummond,
LSOM, ISJ and JLS participated in the study design; LSOM, JMS, and G.R., Dheen, S.T., Sobey, C.G., Jo, D.-G., Chen, C.L.-H., Arumugam, T.V., 2017.
Evidence that NF-κB and MAPK signaling promotes NLRP inflammasome activation
SMSC performed the selection of patients and collection of samples; LSOM, in neurons following ischemic stroke. Mol. Neurobiol. 1–15. https://doi.org/10.
JMS, CMK, and LDC performed the experiments; LSOM and ISJ interpreted 1007/s12035-017-0394-9.
the results; LSOM, ISJ, LDC, DGA, and JAPSM prepared this manuscript. Fernández-Figueroa, E.A., Rangel-Escareño, C., Espinosa-Mateos, V., Carrillo-Sánches, K.,
Salaiza-Suazo, N., Al, E., 2012. Disease severity in patients infected with leishmania
mexicana relates to IL-1 b. PLoS Neglected Trop. Dis. 6. https://doi.org/10.1371/
Funding journal.pntd.0001533.
França-Costa, J., Van Weyenbergh, J., Boaventura, V.S., Luz, N.F., Malta-Santos, H.,
This work was supported by both the Fundação de Amparo à Oliveira, M.C.S., De Campos, D.C.S., Saldanha, A.C., Dos-Santos, W.L.C., Bozza, P.T.,
Barral-Netto, M., Barral, A., Costa, J.M., Borges, V.M., 2015. Arginase I, polyamine,
Pesquisa do Estado da Bahia (FABESB Grant term PET0033/2013) and and prostaglandin E2pathways suppress the inflammatory response and contribute to
the Universidade Estadual de Santa Cruz (UESC).

8
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

diffuse cutaneous leishmaniasis. J. Infect. Dis. 211, 426–435. https://doi.org/10. Howrylak, J., Ryter, S.W., Nakahira, K., Choi, A.M.K., 2015. UCP2-induced fatty acid
1093/infdis/jiu455. synthase promotes NLRP3 inflammasome activation during sepsis. J. Clin. Invest.
Gomes, C., Ávila, L.R., Pinto, S.A., Duarte, F.B., Pereira, L.I.A., Abrahamsohn, I.A., Dorta, 125, 665–680. https://doi.org/10.1172/JCI78253.
M.L., Vieira, L.Q., Ribeiro-Dias, F., Oliveira, M.A.P., 2014. Leishmania braziliensis Mortimer, L., Moreau, F., MacDonald, J.A., Chadee, K., 2016. NLRP3 inflammasome in-
amastigotes stimulate production of IL-1β, IL-6, IL-10 and TGF-β by PBMC from non- hibition is disrupted in a group of auto-inflammatory disease CAPS mutations. Nat.
endemic area healthy residents. Parasite Immunol. 36, 225–231. https://doi.org/10. Immunol. 17, 1176–1186. https://doi.org/10.1038/ni.3538.
1111/pim.12109. Novais, F.O., Carvalho, A.M., Clark, M.L., Carvalho, L.P., Beiting, D.P., Brodsky, I.E.,
Guimarães, E.T., Santos, L.A., Ribeiro dos Santos, R., Teixeira, M.M., dos Santos, W.L.C., Carvalho, E.M., Scott, P., 2017. CD8+ T cell cytotoxicity mediates pathology in the
Soares, M.B.P., 2006. Role of interleukin-4 and prostaglandin E2 in Leishmania skin by inflammasome activation and IL-1β production. PLoS Pathog. 13, e1006196.
amazonensis infection of BALB/c mice. Microb. Infect. 8, 1219–1226. https://doi. https://doi.org/10.1371/journal.ppat.1006196.
org/10.1016/j.micinf.2005.11.011. Nunes, S., Silva, I.B., Ampuero, M.R., Lopes, A.L., 2018. Integrated analysis reveals that
Gupta, A.K., Ghosh, K., Palit, S., Barua, J., Das, P.K., Ukil, A., 2017. Leishmania donovani miR-193b, miR-671, and TREM-1 correlate with a good response to treatment of
inhibits inflammasome-dependent macrophage activation by exploiting the negative human localized cutaneous leishmaniasis. Caused by Leishmania braziliensis 9, 1–13.
regulatory proteins A20 and UCP2. Faseb. J. 31, 5087–5101. https://doi.org/10. https://doi.org/10.3389/fimmu.2018.00640.
1096/fj.201700407R. Osawa, R., Williams, K.L., Singh, N., 2011. The inflammasome regulatory pathway and
Gurung, P., Kanneganti, T.D., 2016. Immune responses against protozoan parasites: a infections: role in pathophysiology and clinical implications. J. Infect. 62, 119–129.
focus on the emerging role of Nod-like receptors. Cell. Mol. Life Sci. 73, 3035–3051. https://doi.org/10.1016/j.jinf.2010.10.002.
https://doi.org/10.1007/s00018-016-2212-3. Paduano, F., Dattilo, V., Narciso, D., Bilotta, A., Gaudio, E., Menniti, M., Agosti, V.,
Gurung, P., Karki, R., Vogel, P., Watanabe, M., Bix, M., Lamkanfi, M., Kanneganti, T., Palmieri, C., Perrotti, N., Fusco, A., Iuliano, R., 2012. Protein tyrosine phosphatase
2015. An NLRP3 inflammasome – triggered Th2-biased adaptive immune response PTPRJ is negatively regulated by microRNA-328. FESB J 280, 401–412. https://doi.
promotes leishmaniasis. J. Clin. Invest. 125, 1329–1338. https://doi.org/10.1172/ org/10.1111/j.1742-4658.2012.08624.x.
JCI79526DS1. Park, H., Huang, X., Lu, C., Cairo, M.S., Zhou, X., 2015. MicroRNA-146a and microRNA-
Haneklaus, M., Gerlic, M., Kurowska-Stolarska, M., Rainey, A.-A., Pich, D., McInnes, I.B., 146b regulate human dendritic cell apoptosis and cytokine production by targeting
Hammerschmidt, W., O'Neill, L.A.J., Masters, S.L., 2012. Cutting edge: miR-223 and TRAF6 and IRAK1 proteins. J. Biol. Chem. 290, 2831–2841. https://doi.org/10.
EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1 production. J. 1074/jbc.M114.591420.
Immunol. 189, 3795–3799. https://doi.org/10.4049/jimmunol.1200312. Peniche, A.G., Bonilla, D.L., Palma, G.I., Melby, P.C., Travi, L., Osorio, E.Y., 2017. A
Hashemi, N., Sharifi, M., Masjedi, M., Tolouei, S., Hejazi, S.H., 2018. Locked nucleic acid secondary wave of neutrophil infiltration causes necrosis and ulceration in lesions of
-anti- let-7a induces apoptosis and necrosis in macrophages infected with Leishmania experimental American cutaneous leishmaniasis. PloS One 12, 1–20. https://doi.org/
major. Microb. Pathog. 119, 193–199. https://doi.org/10.1016/j.micpath.2018.03. 10.1371/journal.pone.0179084. https://search.crossref.org/?q=A+secondary
057. +wave+of+neutrophil+infiltration+causes+necrosis+and+ulceration+in
He, L., Hannon, G.J., 2004. MicroRNAs: small RNAs with a big role in gene regulation. +lesions+of+experimental+American+cutaneous+leishmaniasis.
Nat. Rev. Genet. 5, 522–531. https://doi.org/10.1038/nrg1379. Shio, M.T., Christian, J.G., Jung, J.Y., Chang, K.P., Olivier, M., 2015. PKC/ROS-mediated
Herwaldt, B.L., 1999. Leishmaniasis. Lancet 354, 1191–1199. https://doi.org/10.1016/ NLRP3 inflammasome activation is attenuated by Leishmania zinc-metalloprotease
S0140-6736(98)10178-2. during infection. PLoS Negl. Trop. Dis. 9 (6), e0003868. https://doi.org/10.1371/
Kaye, P., Scott, P., 2011. Leishmaniasis : complexity at the host – pathogen interface. Nat. journal.pntd.0003868.
Publ. Gr. 9, 604–615. https://doi.org/10.1038/nrmicro2608. SINAN, SUVISA, 2016. SITUAÇÃO EPIDEMIOLÓGICA DA LEISHMANIOSE T E G U M E N
Kedzierski, L., Evans, K.J., 2014. Immune responses during cutaneous and visceral TA R A M E R I C A N A (LTA) . B A H I A , 2 0 1 6. Secr. Saúde do Estado da Bahia.
leishmaniasis. Parasitology 141, 1544–1562. https://doi.org/10.1017/ SisLeish-Opas/Who, 2018. LEISHMANIASIS: Epidemiological Report of the Americas.
S003118201400095X. Sutterwala, F., Haasken, S., Cassel, S., 2014. Mechanism of NLRP3 inflammasome acti-
Lamkanfi, M., Dixit, V.M., 2012. Inflammasomes and their roles in health and disease. vation Fayyaz. Ann. N. Y. Acad. Sci. 1319, 82–95. https://doi.org/10.1111/nyas.
Annu. Rev. Cell Dev. Biol. 28, 137–161. https://doi.org/10.1146/annurev-cellbio- 12458.Mechanism.
101011-155745. Tay, H.L., Kaiko, G.E., Plank, M., Li, J., Maltby, S., 2015. Antagonism of miR-328 in-
Lemaire, J., Mkannez, G., Guerfali, F., Gustin, C., Attia, H., Sysco-Consortium, Laouini, D., creases the antimicrobial function of macrophages and neutrophils and rapid clear-
Sghaier, R.M., Dellagi, K., Renard, P., 2013. MicroRNA expression profile in human ance of non- typeable Haemophilus influenzae (NTHi) from infected lung. PLoS
macrophages in response to leishmania major infection. PLoS Neglected Trop. Dis. 7. Pathog. 1–21. https://doi.org/10.1371/journal.ppat.1004549.
https://doi.org/10.1371/journal.pntd.0002478. Tiwari, N., Kumar, V., Gedda, M.R., Singh, A.K., Singh, V.K., Singh, S.P., Singh, R.K.,
Liang, Y., Ridzon, D., Wong, L., Chen, C., 2007. Characterization of microRNA expression 2017. Identification and characterization of miRNAs in response to leishmania do-
profiles in normal human tissues. BMC Genom. 8, 1–20. https://doi.org/10.1186/ novani infection: delineation of their roles in macrophage dysfunction. Front.
1471-2164-8-166. Microbiol. 8, 1–11. https://doi.org/10.3389/fmicb.2017.01190.
Lima-Junior, D.S., Costa, D.L., Carregaro, V., Cunha, L.D., Silva, A.L.N., Mineo, T.W.P., Wang, Y., Wang, Q., Song, J., 2017. Inhibition of autophagy potentiates the proliferation
Gutierrez, F.R.S., Bellio, M., Bortoluci, K.R., Flavell, R. a, Bozza, M.T., Silva, J.S., inhibition activity of microRNA - 7 in human hepatocellular carcinoma cells
Zamboni, D.S., 2013. Inflammasome-derived IL-1β production induces nitric oxide- 3566–3572. https://doi.org/10.3892/ol.2017.6573.
mediated resistance to Leishmania. Nat. Med. 19, 909–915. https://doi.org/10.1038/ Who, 2018. Media centre leishmaniases. http://www.who.int/mediacentre/factsheets/
nm.3221. fs375/en/ accessed 8.20.03.
Liu, C., Zhang, L., Huang, Y., Lu, K., Tao, T., Chen, S., Zhang, X., Guan, H., Chen, M., Xu, Xiao, C., Rajewsky, K., 2009. MicroRNA control in the immune system: basic principles.
B., 2015b. MicroRNA-328 directly targets p21-activated protein kinase 6 inhibiting Cell 136, 26–36. https://doi.org/10.1016/j.cell.2008.12.027.
prostate cancer proliferation and enhancing docetaxel sensitivity. Mol. Med. Rep. 12, Yang, S.F., Lee, W.J., Tan, P., Tang, C.H., Hsiao, M., Hsieh, F.K., Chien, M.H., 2015a.
7389–7395. https://doi.org/10.3892/mmr.2015.4390. Upregulation of miR-328 and inhibition of CREB-DNA-binding activity are critical for
Liu, W., Zhang, Y., Xia, P., Li, S., Feng, X., Gao, Y., Wang, K., Song, Y., Duan, Z., Yang, S., resveratrol-mediated suppression of matrix metalloproteinase-2 and subsequent
Shao, Z., Yang, C., 2016. MicroRNA-7 regulates IL-1β-induced extracellular matrix metastatic ability in human osteosarcomas. Oncotarget 6, 2736–2753. https://doi.
degeneration by targeting GDF5 in human nucleus pulposus cells. Biomed. org/10.18632/oncotarget.3088.
Pharmacother. 83, 1414–1421. https://doi.org/10.1016/j.biopha.2016.08.062. Yang, Z., Zhong, L., Xian, R., Yuan, B., 2015b. MicroRNA-223 regulates inflammation and
Liu, Y., Zhang, X., Zhang, Y., Hu, Z., Yang, D., Wang, C., Guo, M., Cai, Q., 2015a. brain injury via feedback to NLRP3 inflammasome after intracerebral hemorrhage.
Identification of miRNomes in human stomach and gastric carcinoma reveals miR- Mol. Immunol. 65, 267–276. https://doi.org/10.1016/j.molimm.2014.12.018.
133b/a-3p as therapeutic target for gastric cancer. Canc. Lett. 369, 58–66. https:// Zhou, Y., Lu, M., Du, R., Qiao, C., Jiang, C., Zhang, K., Ding, J., 2016. MicroRNA-7 targets
doi.org/10.1016/j.canlet.2015.06.028. Nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the
Maretti-Mira, A., Oliveira-Neto, M.P., Da-Cruz, A.M., Craft, N., Pirmez, C., 2010. pathogenesis of Parkinson ’ s disease. Mol. Neurodegener. 1–15. https://doi.org/10.
Therapeutic failure in American cutaneous leishmaniasis is associated with gelatinase 1186/s13024-016-0094-3.
activity and cytokine expression. Clin. Exp. Immunol. 163, 207–214. https://doi.org/
10.1111/j.1365-2249.2010.04285.x.
Marques-Rocha, J.L., Samblas, M., Milagro, F.I., Bressan, J., Martínez, J.A., Marti, A., Lucilla Silva Oliveira Mendonça. Master's Degree student
2015. Noncoding RNAs, cytokines, and inflammation-related diseases. Faseb. J. 29, at the Universidade Estadual de Santa Cruz (Ilhéus-BA-
3595–3611. https://doi.org/10.1096/fj.14-260323. Brazil) from 2016 to 2018. She was part of the “Role of
Martinon, F., Burns, K., Tschopp, J., 2002. The Inflammasome: a molecular platform Inflammasomes in Cutaneous Leishmaniasis” research
triggering activation of inflammatory caspases and processing of proIL-β. Mol. Cell. group and currently is PhD student in the same institution
10, 417–426. https://doi.org/10.1016/S1097-2765(02)00599-3. from 2018 to 2022, acting on a project related to im-
Martinon, F., Tschopp, J., 2005. NLRs join TLRs as innate sensors of pathogens. Trends munomodulation of human phagocytes.
Immunol. 26, 447–454. https://doi.org/10.1016/j.it.2005.06.004.
Matte, C., Maion, G., Mourad, W., Olivier, M., 2001. Leishmania donovani -induced
macrophages cyclooxygenase-2 and prostaglandin E 2 synthesis. Parasite Immunol.
23, 177–184. https://doi.org/10.1046/j.1365-3024.2001.00372.x.
Miska, E.A., 2005. How microRNAs control cell division, differentiation and death. Curr.
Opin. Genet. Dev. 15, 563–568. https://doi.org/10.1016/j.gde.2005.08.005.
Moon, J.S., Lee, S., Park, M.A., Siempos, I.I., Haslip, M., Lee, P.J., Yun, M., Kim, C.K.,

9
L.S.O. Mendonça, et al. Experimental Parasitology 210 (2020) 107846

Jaqueline Marques dos Santos. Graduated in Nursing at Danillo Gardenal Augusto. Bachelor in Biological Sciences
the State University of Santa Cruz in the year 2013, com- (2006), Master in Genetics (2008) and PhD in Genetics
pleting it in 2018. During this period, she was a student of awarded by Federal University of Paraná. Brazil. (2012).
Scientific Initiation at the Laboratory of Immunobiology, Postdoctoral experience in prestigious institutions such as
where she developed the project entitled: Role of Harvard Medical School (USA) and the National Institutes
Inflamamosomes and Polymorphisms Genetics in of Health (USA). Experienced in Genetics and Immunology,
Resistance and Susceptibility to American Cutaneous with emphasis on human molecular genetics, especially in
Leishmaniasis. He is currently a student of the Graduate immunogenetics, population genetics, KIR and HLA genes,
Program in Immunology of the University of São Paulo, genetic susceptibility to complex and infectious diseases,
developing his research project in the Immunology molecular biology, gene expression and next generation
Laboratory of Mucosa, studying more specifically the mu- sequencing. Currently holds a position of Full Specialist at
cosa of the gastrointestinal tract. the University of California, San Francisco.

Dr. Carla M. Kaneto is a professor of Human Genetics at Silvia Maria de Carvalho. Bachelor in Biological Sciences
Santa Cruz State University. She has coauthored publica- at the Catholic University of Salvador; Master in Genetics at
tions including stem cell differentiation and gene expres- the Federal University of Pernambuco; PhD in Public Health
sion in Cancer, Mesenchymal Stem Cells and Cardiovascular at the Ageu Magalhães Research Center, FIOCRUZ-PE.
Diseases. The current research interests in Professor Currently working as a Professor of Human Parasitology,
Kaneto's group include MicroRNAs expression in cardio- Medical Parasitology and Medical Entomology at State
vascular diseases, diabetes and leukemia. University of Santa Cruz. Experienced in Leishmaniasis and
enteric parasites.

Jamária A. P. Soares Martins. Dr. Martins graduated in


Biological Sciences at Universidade Federal de Viçosa in
Dr Luciana Debortoli de Carvalho is a visiting professor 2002, where she started working with microbiology. She
in the area of microbiology and immunology at the Santa earned her Master (2004) and Ph.D. (2007) both in
Cruz State University. I have experience in Microbiology Microbiology at Universidade Federal de Minas Gerais, still
working mainly in the following subjects: Immunology, in Brazil. While working there, Dr. Martins' research helped
Virology, Bacteriology, General and Medical Mycology, to elucidate critical differences in the cellular signaling
Clinical Analyzes, Molecular biology and bioprospecting of pathways induced by genetically related poxviruses,
plants with biological activity against microorganisms. leading to viral tropism. Dr. Martins completed her
Postdoctoral Fellowship at The Medical College of
Wisconsin in Milwaukee, USA (2013). She currently teaches
science classes at the technical colleges (both MATC and
WCTC) in Milwaukee area.

Izaltina Silva-Jardim. Bachelor in Biological Sciences at


Jane Lima dos Santos. PhD in Biochemistry and the Federal University of Minas Gerais, Master in Basic and
Immunology, MA in Microbiology, and BSc in Biological Applied Immunology and PhD degree in Biochemistry
Science. Currently working as a research/professor at awarded by Ribeirão Preto Medical School, University of
Universidade Estadual de Santa Cruz (UESC), Ilhéus/BA. São Paulo. From 2005 to 2010, she was a researcher at
Some areas of research include cellular immunology, im- Institute of Research in Tropical Disease – IPEPATRO
munity to microorganisms with emphasis on host-micro- (current FIOCRUZ – Rondônia) and a visitant professor at
organism interaction In vitro and In vivo, and the impact of Federal University of Rondônia. Postdoctoral experience at
biofungicide and biofertilizers in this interaction. Advisor São Carlos Institute of Physics, University of São Paulo.
to master degree and PhD students connected to UESC Currently working as a professor/researcher at State
Biology and Biotechnology of Microorganism University of Santa Cruz –UESC. Her research focuses in
Postgraduation program. Engaged in university extension chemotherapy for leishmaniasis and host-parasite interac-
activities, such as projects to disseminate and popularize tion.
Science in the area of health knowledge.

10

You might also like