Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Acta Parasitologica

https://doi.org/10.2478/s11686-020-00188-0

ORIGINAL PAPER

Zinc Supplementation: Immune Balance of Pregnancy During


the Chronic Phase of the Chagas Disease
Cássia Mariana Bronzon da Costa1 · Marina del Vecchio Filipin1 · Fabrícia Helena Santello1 ·
Inara Fernanda Lage Gallo1 · Luiz Miguel Pereira1 · Fernando Barbosa Jr1 · José Clóvis do Prado Júnior1 ·
Ana Amélia Carraro Abrahão1

Received: 10 June 2019 / Accepted: 18 February 2020


© Witold Stefański Institute of Parasitology, Polish Academy of Sciences 2020

Abstract
Background  Chagas disease or American trypanosomiasis is caused by the protozoan Trypanosoma cruzi and is endemic of
the Americas. The control of the disease is restricted to toxic and potentially teratogenic drugs, which limit the use during
pregnancy. The use of food supplementation offers a safe and low-cost form to alleviate Chagas disease symptoms, mostly
in areas with alimentary risk. For example, zinc demonstrates positive effects in immune response, including in Chagas
disease during pregnancy.
Purpose  This study describes the innate response in pregnant rats chronically infected with T. cruzi and supplemented with
zinc.
Methods  Pregnant female Wistar rats, infected with T. cruzi, were treated with 20 mg/kg/day zinc sulfate and euthanized on
the 18th day. Samples (plasma, splenocytes, and peritoneal exudate) were collected and several immune parameters (nitric
oxide, RT1B, CD80/CD86, MCP-1, CD11b/c, NK/NKT, IL-2, IL-10, INF-cc, and apoptosis) evaluated.
Results  Under Zinc supplementation and/or T. cruzi infection, the gestation developed normally. Several innate immune
parameters such as RT1B, CD80/CD86, MCP-1 expressing lymphocytes, IL-2, and IL-17 were positively altered, whereas
nitric oxide, CD11b/c, NK/NKT, apoptosis, INF-γ, and corticosterone demonstrated a pro-pregnancy pattern.
Conclusion  Our results indicated that zinc has diverse effects on immune response during pregnancy. An anti-T. cruzi
immunity, as well as a pro-gestation response, were observed after zinc supplementation. The complete comprehension of
zinc supplementation in pregnancy will base an adequate strategy to alleviate Chagas disease symptoms and propagation,
especially for populations from endemic areas.

Keywords  Chagas disease · Pregnancy · Zinc · Chronic phase

Introduction

Trypanosoma cruzi is the causative agent of Chagas disease,


which afflicts millions of people of America. The disease is
transmitted after a bite and contact with contaminated feces
Electronic supplementary material  The online version of this
article (https​://doi.org/10.2478/s1168​6-020-00188​-0) contains of Triatomine insects. Contaminated insect feces also infect
supplementary material, which is available to authorized users. food and lead to oral transmission. Blood transfusion, organ
transplantation, and congenital transmission are related to
* Cássia Mariana Bronzon da Costa the expansion of the Chagas disease to non-endemic regions,
marianabronzon@gmail.com
such as North America, Europe, Japan, and Australia [53].
* Ana Amélia Carraro Abrahão Due to the importance of congenital transmission, the
acarraro@fcfrp.usp.br
dynamic of Chagas disease during pregnancy is a key point
1
Department of Clinical Analysis, Toxicology and Food to describe and control the parasite propagation. Moreover,
Science, School of Pharmaceutical Sciences of Ribeirão the classic anti-T. cruzi drugs (benznidazole and nifurtimox)
Preto, University of São Paulo, Ribeirão Preto 14040‑903, have been related to toxicity and/or teratogenicity [11, 39].
Brazil

13
Vol.:(0123456789)
Acta Parasitologica

Thus, novel strategies are necessary to alleviate the effects non-treated pregnant animals (PC), non-infected/zinc-treated
of the disease in pregnancy. pregnant animals (PCZ), T. cruzi-infected/non-treated preg-
The gestation is characterized by genetic and hormonal nant animals (PI), T. cruzi-infected/zinc-treated pregnant
alterations, to continually provide nutrients to the embry- animals (PIZ).
onic development. During the gestational period, a maternal
immune tolerance is developed to accept the fetus [43]. The
metabolism is also altered during pregnancy, and an ade- Pregnancy and T. cruzi Infection
quate caption of nutrients is necessary. Imbalances in nutri-
tion during pregnancy represent a constant risk for mother The animals were infected intraperitoneally with 1 × 105 try-
and fetus. According to the World Health Organization [53], pomastigotes forms of T. cruzi Y strain [42] and housed for
the adequate intake of nutrients is one of the determinants 30 days. This period of infection (transition of the acute to
of fetal growth, with repercussions on the weight and gesta- the chronic phase of Chagas disease) is characterized by the
tional age at birth. Therefore, among the diverse nutrients, decrease of trypomastigotes counting in peripheral blood
zinc has an important role in the immune response and has [28, 32]. On the 30th day after infection (when no trypomas-
been related to efficient control of viral, bacterial, and pro- tigotes were observed in peripheral blood), the female rats
tozoal infections [9]. Our group has studied the effects of were maintained with male counterparts, in a proportion of
zinc supplementation during the pregnancy in rats infected 2:1 per cage and the pregnancy was confirmed by the pres-
with T. cruzi [12–14]. Zinc has a fundamental role in growth, ence of the vaginal plug [12].
development and differentiation of tissues, and embryogen-
esis [22, 36, 49]. Moreover, clinical trials have demonstrated
that maternal zinc deprivation increases the risk of mortality, Zinc Supplementation and Euthanasia
growth retardation, and fetal malformations, including neu-
ral tube defects [25]. Zinc stimulates the immune response in On the first day of pregnancy, animals were treated with
the acute phase of the Chagas disease, and several adaptive 20 mg/kg/day zinc sulfate (Sigma-Aldrich, Catalog number:
parameters were improved in the chronic phase [12, 14]. In 1724769) by oral gavage for 18 days [12]. At the final stage
this study, we described the innate immune response during of the pregnancy (18th day), the animals were anesthetized
the chronic phase of Chagas disease and the role of the zinc with 2.5% tribromoethanol and euthanized by decapitation.
supplementation. Zinc improved several innate immune-
response parameters (RT1B, CD80/CD86, MCP-1, IL-2, and
IL-17). In parallel, other points indicated a pro-gestation pat- Fetuses and Placentas Analysis
tern (nitric oxide, CD11b/c, NK/NKT, apoptosis, INF-γ, and
corticosterone), balancing the pro-inflammatory response After euthanasia, the fetuses and placentas were collected
induced against the infection. Therefore, zinc promotes the and the weight and length/diameter measured using an ana-
immune response without compromising fetal development. lytic balance (Sartorius, Goettingen, Germany) and a digital
Although the results are promising, future assays will be caliper (Mitutoyo, Kawasaki, Japan), respectively.
necessary to elucidate the mechanisms related to the zinc
supplementation during pregnancy associated with the Cha-
gas disease. Adequate nutrient intake is fundamental for the Quantification of Serum Zinc
development and health of the mother and fetus. Special
attention may be directed to Chagas disease-endemic areas, Zinc quantification was adapted from a method previously
where a food risk is also considered. described [3] with little modification for rat serum sam-
ples analysis [13]. Briefly, serum was diluted (1:20) with a
solution containing 0.01% (v/v) T ­ riton® X-100, 0.5% (v/v)
Material and Methods −1
nitric acid, and 10 µg ­L of each one of Rh (as internal
standard). Analyses were carried out with an inductively
Animals coupled plasma mass spectrometer equipped with a reaction
cell (ICP-MS ELAN DRCII, PerkinElmer, SCIEX, Norwalk,
Female Wistar rats (five animals/group) were used and CT) operating with high-purity argon (99.999; Praxaair, Bra-
housed in the Facility House of the São Paulo University zil). The sample introduction system was composed of a
Campus of Ribeirão Preto, with water and food at libitum. quartz cyclonic spray chamber and a M ­ einhard® nebulizer
®
The animal manipulation protocol was approved by the connected by ­Tygon tubes to the ICP-MS’s peristaltic pump
local Ethics Committee (Protocol number 11.1.1210.53.2). (set at 20 rpm). The ICP-MS was operated with Pt sampler
Four experimental groups were designed: non-infected/ and skimmer cones purchased from Perkin Elmer.

13
Acta Parasitologica

Peritoneal and Splenic Cells The data acquisition was performed in a FACS Canto flow
cytometer (BD Biosciences, California, USA) equipped with
The peritoneal cells were collected by injection of cold the FACSDiva software (BD Biosciences, California, USA).
RPMI 1640 medium (10 mL, Sigma-Aldrich, USA) in the
peritoneal cavity of animals. The suspension was centrifuged T. cruzi Protein Extract
at 410g for 15 min and the pellet suspended in 2 mL of
RPMI 1640 medium. The splenic cells were harvested by the For evaluation of the apoptosis under specific stimulation,
mechanical disaggregation of spleen followed by a passage T. cruzi protein extracts were used. Trypomastigote forms
in a 70-μm nylon cell strainer (Falcon, USA). The disaggre- (Y strain) were cultivated in cell cultures (LLCMK2 line-
gated cells were washed with a hypotonic buffer (160 mM age). The culture supernatants were collected and filtered in
NH4Cl, 10 mM Tris–HCl, pH 7.4) and suspended in RPMI a 5 μm filter syringe to separate cells and trypomastigotes.
1640. The peritoneal and splenic cells were counted in a The parasites (1 × 108/mL) were washed with PBS and sus-
hemocytometer, and the cellular viability verified after dilu- pended in RPMI. The suspension was sonicated (two soni-
tion (1:2) in a 0.4% trypan blue solution (Sigma-Aldrich cation cycles at 20 kHz, 30 W in a QSonic sonicator, Cole
catalog number: T8154). For general use, the cells were Parmer, USA), centrifuged for 10 min, at 10,000g, 4 °C and
diluted to 2 × 106 cells/mL. the supernatant filtered in 0.22 μm filter syringe. The T. cruzi
protein extract was quantified by the Bradford protein assay
and diluted to 3 mg/mL.
Nitric Oxide Quantification
Intracellular MCP‑1 Detection
Peritoneal cells were distributed (2 × 105/well) in 96-well
plates (TPP, Switzerland) and incubated with 1  μg/mL Splenocytes were distributed in 96-well plates (Corning Inc.,
lipopolysaccharide (LPS, Sigma-Aldrich, catalog num- USA) and incubated with 500 ng/mL ionomycin (Sigma-
ber: L2654) for 48 h, 37 °C, 5% ­CO2. After incubation, the Aldrich, USA) and 50 ng/mL phorbol 12-myristate 13-ace-
plates were centrifuged at 1500g, for 4 min, 20 °C and the tate (PMA, Sigma-Aldrich, USA) for 4 h at 37 °C in 5%
supernatant (50 μL/well) transferred to a new 96-well plates. ­CO2. After incubation, cells were cultured with brefeldin
The nitrite measurement was performed according to the A (10 μg/mL, BD Pharmingen) for 2 h, 37 °C in 5% C ­ O 2,
Griess reaction protocol [5]. The samples were incubated washed in Stain Buffer BSA (BD Pharmingen) and blocked
with 100 μL of Griess solution (1% sulfanilamide, 0.1% with Fc block (anti-rat CD32, BD Pharmingen). Surface
N-(1-Naphthyl) ethylenediamine in 5% phosphoric acid) for staining was performed with anti-rat CD3/FITC, anti-rat
15 min at room temperature and the plates read in an ELISA CD4/PECy7, and anti-rat CD8/PerCP (BD Pharmingen).
reader at 540 nm (Synergy H1, Biotek, USA). The nitrite After surface staining, cells were fixed and permeabilized
concentrations were calculated by linear regression using a with Cytofix/Cytoperm solution (BD Pharmingen), washed
sodium nitrite curve (500–7.8 μM, serially diluted). with Perm/Wash solution (BD Pharmingen) and intracel-
lularly labeled with anti-rat MCP-1/PE. The labeled cells
were washed in Perm/Wash solution, fixed in 1% paraform-
Phenotypic Analysis of Peritoneal and Splenic Cells aldehyde in PBS and analyzed by flow cytometry in a FACS
Canto flow cytometer (Becton Dickinson Immunocytometry
A similar protocol was adopted for phenotypic analysis of Systems), using FACSDiva software.
peritoneal and splenic cells, according to [12, 14]. Briefly,
2 × 106 cells in 12 × 75 mm round-bottomed polystyrene Annexin V and Propidium Iodide
tubes (Falcon, USA) were fixed with 3.5% paraformalde-
hyde for 15 min and washed with phosphate-buffered saline For early and late apoptosis, splenic cells (2 × 105/well)
(PBS). The cells were suspended in Fc receptor block- were distributed in 96-well plates in RPMI 1640. The
ing (anti-CD32, BD Pharmingen) for 20 min, at 4 °C and cells were incubated for 24  h, at 37  °C, 5% ­C O 2 with
washed with PBS. The samples were suspended in Cell 0.3 mg/mL T. cruzi protein extract or RPMI 1640 (con-
Staining Buffer (BD Pharmingen) and labeled with anti-rat trol). In parallel, cells were incubated under the same
RT1B/PercP, anti-rat CD11bc/PECy7, anti-rat CD86/FITC, conditions for 15 min, however, without T. cruzi protein
anti-rat CD80/PE (peritoneal cells) or anti-rat CD3/APC, extract. The cells were transferred to 12 × 75 mm round-
anti-rat CD161-/FITC (splenic cells) for 30 min, at 4 °C. The bottomed polystyrene tubes, centrifuged for 5 min, 2500g,
cells were washed with PBS and suspended in Cell Stain- 4 °C and washed with PBS. The samples were suspended
ing Buffer for flow cytometry analysis. All antibodies were in 1:100 Annexin V-FITC diluted in binding buffer (BD
purchased from BD Biosciences Pharmingen, CA, USA. Biosciences Pharmingen) and incubated for 15  min,

13
Acta Parasitologica

at room temperature in the dark. Before the cytometry Results


analysis, propidium iodide (PI, BD Biosciences Pharmin-
gen) was added to the suspensions. The cells under early Zinc Serum Levels and Effects on Fetuses
(Annexin ­V + ­P I −) and late apoptosis (Annexin V
­ + ­P I +) and Placentas
were detected after analysis in a FACS Canto flow cytom-
eter (BD Biosciences, California, USA) equipped with the The T. cruzi infection and/or the zinc supplementation did
FACSDiva software. not alter the weight and length/diameter of fetuses and pla-
centas, compared to the non-infected and non-treated control
(Suppl. Fig. 1). The zinc serum levels were constant among
Cytokine Quantification groups, and no statistical difference was observed. The zinc
concentrations in sera were 2.8 ± 0.5 μg/mL, 3.5 ± 0.7 μg/
Concentrations of IFN-γ, IL-2, and IL-17 were measured mL, 6.6 ± 3.7 μg/mL, and 5.4 ± 0.9 μg/mL for PC, PCZ, PI,
by specific two-site enzyme-linked immunosorbent assay and PIZ, respectively. (Suppl. Fig. 2).
(ELISA) according to the manufacturer’s specifications.
For quantification, reference standard curves were uti-
Nitric Oxide Production and Antigen Presentation
lized. All kits were purchased from R&D Systems (Min-
neapolis, MN, USA). The samples were processed indi-
During the analyzed period of T. cruzi infection (30th–48th
vidually (five animals/group) and plates read at 450 nm
days post-infection), the nitric oxide production was unal-
in an ELISA reader (Synergy H1, Biotek).
tered among the groups (Suppl. Fig. 3). However, the antigen
presentation was responsive to infection and/or zinc supple-
mentation (Fig. 1). The percentage of CD11b/c+ cells from
Corticosterone Detection
peritoneum decreased after T. cruzi infection (PI), compared
to the non-infected/non-treated group (PC). Although the
The corticosterone levels from plasma samples were
zinc supplementation (PIZ) reverted this condition in rela-
determined using the Corticosterone ELISA kit (Abcam,
tion to the infected group, the proportion of the CD11b/c+
catalog number: 108821). The measurement was per-
cells was lower compared to the PC group (Fig. 1a). The
formed following the manufacturer’s instructions, and
­MHCII+ cells were similar among infected (PI) and non-
the plates read in an ELISA reader (Synergy H1, Biotek)
infected animals (PC or PCZ), whereas the infected and
at 450 nm. The corticosterone levels were quantified in
zinc-treated group (PIZ) demonstrated a significant eleva-
relation to a standard curve provided by the manufacturer.
tion (Fig. 1b).

Statistical Analysis Antigen‑Presenting Cell Costimulation (CD80/CD86)

The values were plotted as mean ± standard deviation (SD). The antigen-presenting cells (CD11b/c +) that express
The groups were analyzed by one-way ANOVA followed by costimulatory proteins (­ CD80 +/CD86 +) were increased
the Tukey post test. All statistical analyses were performed in infected animals treated with zinc (PIZ). The cells
in the GraphPad Prism 5.0 software (La Jolla, CA, USA). (CD11b/c + CD80 + ) were unaltered in infected (PI) or

Fig. 1  Antigen cell presentation. Female Wistar rats were infected anized. The peritoneal cells were collected, processed, labeled with
with T. cruzi and after 30 days submitted to mating. After pregnancy anti-rat CD11b/c or anti-rat RT1B, and analyzed by flow cytometry. a
confirmation, the animals were treated with zinc for 18 days and euth- CD11b/c+ cells. b ­RT1B+ cells. *p < 0.05

13
Acta Parasitologica

non-infected/zinc-treated (PCZ) animals, whereas were Detection of MCP‑1‑Expressing Lymphocytes


significantly elevated in the infected and zinc-treated
group (PIZ) compared to the control (PC) (Fig. 2a). The The MCP-1-expressing lymphocytes (­CD4 + or C ­ D8 +)
(CD11b/c+CD86+) cells from infected animals (PI) were were elevated under T. cruzi infection in relation to the
decreased in relation to the control (PC). When ani- non-infected/non-treated group (PC). For ­CD4+MCP-1+
mals were zinc treated (PIZ), the proportion of (CD11b/ cells, the zinc supplementation (PIZ) increased the MCP-
c+CD86+) cells was elevated compared to infected or con- 1-expressing cells compared to the infected animals (PI)
trol groups (Fig. 2b). (Fig.  4a), whereas the ­CD8+MCP-1+ lymphocytes were
unaltered (Fig. 4b).

Detection of NK ­(CD161+) and NKT ­(CD3+CD161+) Apoptosis


Cells
The early and late apoptosis of cells incubated for 15 min
The splenic cells labeled with anti-rat CD161 were unal- demonstrated the opposite patterns. Whilst the early apop-
tered among all groups (Fig. 3a). However, NKT cells tosis was unaltered after T. cruzi infection and/or zinc sup-
­(CD3+CD161+) were responsive to T. cruzi infection and plementation; the late apoptosis was reduced in relation to
zinc supplementation. The T. cruzi infection increased the the control (C) (Fig. 5a, b; Suppl. Fig. 4). The infection with
­CD3+CD161+ cells when compared to the control (PC). T. cruzi decreased the early and late apoptosis in cells incu-
The zinc supplementation (PIZ) reversed this pattern bated for 24 h with T. cruzi extract or RPMI. When supple-
by decreasing the C ­ D3 +CD161 + cells to control levels mented with zinc, the apoptosis (early or late) from infected
(Fig. 3b). animals was increased, however, only in cells incubated

Fig. 2  CD80+/CD86+-expressing cells. Female Wistar rats were cessed, and labeled with anti-rat CD11b/c. The CD11b/c+ cells were
infected with T. cruzi and after 30  days submitted to mating. After also labeled with anti-rat CD80 or anti-rat CD86 and analyzed by
pregnancy confirmation, the animals were treated with zinc for flow cytometry. a CD11b/c+CD80+ cells. b CD11b/c+CD86+ cells.
18  days and euthanized. The peritoneal cells were collected, pro- *p < 0.05

Fig. 3  NK and NKT cells. Female Wistar rats were infected with T. nized. The splenic cells were collected, processed and labeled with
cruzi and after 30  days submitted to mating. After pregnancy con- anti-rat ­CD161+ and/or anti-rat ­CD3+ and analyzed by flow cytom-
firmation, the animals were treated with zinc for 18 days and eutha- etry. a ­CD161+ cells. b ­CD3+CD161+ cells. *p < 0.05

13
Acta Parasitologica

Fig. 4  MCP-1 expressing lymphocytes evaluation. Female Wistar anti-rat CD32. The cells were washed and labeled with anti-rat CD3,
rats were infected with T. cruzi and after 30  days submitted to mat- anti-rat CD4, and anti-rat CD8. For the detection of secreted MCP-
ing. After pregnancy confirmation, the animals were treated with 1, the cells were permeabilized, labeled with anti-rat MCP-1 and
zinc for 18 days and euthanized. The splenic cells were collected and analyzed by flow cytometry. a ­CD4+MCP-1+ cells. B ­CD8+MCP-1+
incubated with ionomycin and PMA for cell secretion. The secretion cells. *p < 0.05
process was blocked with brefeldin A and the cells incubated with

for 24 h (under T. cruzi stimulation); the early apoptosis of the pathogen and/or the treatment. Thus, immunomodu-
was superior concerning the control (C) (Fig. 5c–f; Suppl. latory molecules have an interesting potential, which may
Fig. 4). For cells incubated for 24 h, the zinc supplementa- increase the response against pathogens as well as the side
tion in healthy animals (PCZ) maintained apoptosis com- effects caused by drugs. Due to the low zinc toxicity at nor-
pared to the control (C), except the early apoptosis in non- mal intakes (as applied in this study), the element has the
stimulated cells (RPMI) (Fig. 5c; Suppl. Fig. 4). potential for use during pregnancy. As observed, the sup-
plementation does not alter the zinc serum levels, the weight
Cytokine Quantification or length of fetuses and placentas. Moreover, all pregnan-
cies occurred normally, and no fetal malformations were
The infection with T. cruzi (PI) demonstrated distinct pat- detected. Our data indicate that zinc supplementation is safe
terns in the INF-γ, IL-2, and IL-17 levels. The infection for use during pregnancy, even under T. cruzi infection.
elevated INF-γ and IL-17 detection in serum, whereas the The chronic phase of the Chagas disease and/or zinc sup-
IL-2 levels were unaltered compared to the control (C) plementation altered several parameters in pregnant rats.
(Fig. 6). The treatment of infected animals with zinc (PIZ) In this study, one of our aims was directed to antigen-pre-
decreased the INF-γ levels, reaching the control parameters senting cells from peritoneal exudate. The innate response
(C) (Fig. 6a). For IL-2 and IL-17, the treatment of infected induced by T. cruzi infection decreased the dendritic cells
animals (PIZ) increased the levels of these cytokines in rela- as well as the cells expressing MHC II molecules. These
tion to all groups (PC, PCZ, or PI) (Fig. 6b, c). cells are pivotal for the lymphocyte and phagocyte stimula-
tion and T. cruzi down-regulates APCs to evade of immune
Corticosterone Quantification response [21, 50]. Moreover, macrophages and dendritic
cells are important for the maternal–fetal balance, propiti-
The T. cruzi infection decreased the corticosterone levels ating the immunological tolerance as well as the activation
compared to the controls (PC and PCZ), and the pattern was of a particular response against pathogens [2, 31]. The zinc
maintained after zinc supplementation (PIZ) (Fig. 7). supplementation in infected animals increased the propor-
tion of these cells compared to the infected ones. In parallel,
the nitric oxide (NO) is unaltered among groups. Despite the
Discussion effectiveness of NO for parasite control [34, 52], an excess
of this molecule leads to tissue damage, including cardio-
The pregnancy is a special condition in which the mater- myopathy [7]. Indeed, during the conversion of acute to the
nal immune system is regulated to tolerate and allows fetal chronic stage of Chagas disease (20–60 days post-infection),
development [54]. This immune balance reacts differently the parasite proliferation is usually controlled by the immune
to pathogens and exogenous molecules compared to males system and a regulated nitric oxide production is welcomed
and non-pregnant females [31]. The control of Chagas dis- to avoid maternal–fetal disturbance. For an adequate T-cell
ease in pregnant models is limited due to the toxicity of activation and proliferation, the APC’s surface proteins
the benznidazole and nifurtimox [11, 39], which urges the CD80 and CD86 interact with CD28 expressed in most T
necessity for novel forms to alleviate the deleterious effects lymphocytes [10]. The CD80/86-CD28 costimulation has a

13
Acta Parasitologica

Fig. 5  Early and late apoptosis of splenic cells. Female Wistar rats before the analysis by flow cytometry. a Early apoptosis of cells
were infected with T. cruzi and after 30  days submitted to mating. incubated for 15  min in RPMI. b Late apoptosis of cells incubated
After pregnancy confirmation, the animals were treated with zinc for for 15 min in RPMI. c Early apoptosis of cells incubated for 24 h in
18  days and euthanized. The splenic cells were collected, processed RPMI. d Late apoptosis of cells incubated for 24 h in RPMI. e Early
and incubated with T. cruzi extract or RPMI for 15  min or 24  h, apoptosis of cells incubated for 24 h in T. cruzi extract. f Late apopto-
37 °C, 5% ­CO2. The cells were washed and incubated with Annexin sis of cells incubated for 24 h in T. cruzi extract. *p < 0.05
V and propidium iodide (PI) for 15  min and 5  min, respectively,

crucial role in T. cruzi control and animals lacking functional tissues [26, 41]. Studies using Toxoplasma gondii and Myco-
CD28 and/or CD80/86 are highly susceptible to the acute bacterium tuberculosis indicate that the MCP-1 absence
phase of the Chagas disease [29]. However, T-cell prolifera- reduces the ability to control infections [4, 37]. Likewise,
tion is decreased after zinc supplementation in pregnant rats MCP-1 is produced in large quantities in the heart of T.
infected with T. cruzi [14]. Thus, novel assays are necessary cruzi-infected mice to increase parasite destruction. MCP-1
to clarify the relation between CD80/CD86 expression and knockout mice develop higher parasitemia and mortality
T-cell proliferation during pregnancy in animals infected when compared to wild-type (WT) counterparts [33]. Simi-
with T. cruzi. Although zinc decreases the T-cell prolifera- larly, NK and NKT cells contribute to the control of chronic
tion, MCP-1-expressing T lymphocytes are elevated after infections, including Chagas disease [17]. The depletion
T. cruzi infection and zinc supplementation. MCP-1/CCL2 of NK and NKT cells in mice also increases the T. cruzi
chemokine is secreted by a large number of cells in inflam- parasitemia and decreases the survival rate [6, 38]. In preg-
matory processes for monocyte recruitment to infected nancy, high levels of NK cells are related to complications

13
Acta Parasitologica

Fig. 7  Corticosterone plasma levels. Female Wistar rats were infected


with T. cruzi and after 30 days submitted to mating. After pregnancy
confirmation, the animals were treated with zinc for 18  days and
euthanized. The plasmas were collected, and the corticosterone levels
quantified using a Corticosterone ELISA kit (Abcam). *p < 0.05

elevation of apoptosis contributes to the elimination of anti-


paternal T cells, preventing deleterious immune responses
towards the fetus [1, 24]. On the other side, patients with
recurrent spontaneous abortion demonstrate significantly
lower T-cell apoptosis compared to fertile women [20]. Our
results indicate that the T. cruzi infection decreased the cell
apoptosis, mostly when cells were incubated with T. cruzi
antigens. However, the zinc supplementation of infected
animals improved the early and late apoptosis in cells incu-
bated with T. cruzi antigens, compared to the infected and
non-supplemented group. Thus, zinc contributes to balanced
apoptosis, maintaining the immune response as well as the
protection of mother and fetus.
INF-γ, a pro-inflammatory cytokine, is produced by
macrophages, NK, and T cells to stimulate the antigen-pre-
senting cells, NO production, NK activity, and B-cell pro-
liferation [40, 48]. This molecule acts for the control of the
Fig. 6  INF-γ, IL-2, and IL-17 quantification. Female Wistar rats parasite propagation and is also related to cardiac damage
were infected with T. cruzi and after 30  days submitted to mating.
After pregnancy confirmation, the animals were treated with zinc for
[7, 46]. Moreover, INF-γ induces a potent TH1 response,
18  days and euthanized. The sera were collected, and the levels of which leads to fetal loss and preterm labor in the mouse
INF-γ, IL-2, and IL-17 quantified by the Rat Quantikine ELISA Kit model [45]. Therefore, the control of INF-γ production after
(R&D Systems), using specific standard curves. *p < 0.05 zinc supplementation contributes to a pro-pregnancy course.
Otherwise, IL-2 is improved after zinc supplementation indi-
during pregnancy such as preeclampsia and hypertension cating a pro-inflammatory response in pregnancy. Although
[8]. However, the mechanisms concerning NK and NKT IL-2 administration inhibits pregnancy in a mouse model
role in the immune response against T. cruzi or pregnancy [47], this cytokine has an important effect on controlling T.
remain unclear. T. cruzi infection induces T-cell apoptosis cruzi infection. IL-2 released by macrophage stimulates a
as a mechanism to the propagation and immune system eva- protective immune response against T. cruzi, inducing the
sion. Intense apoptosis induced in Chagas disease interferes production of INF-γ by T and NK cells [18]. Once INF-γ
with T-cell activation and production of pro-inflammatory is downregulated in pregnant rats infected with T. cruzi and
cytokines [16]. However, in our model of infection, the Cha- treated with zinc, there is a balance between anti-pathogen
gas disease decreased the percentage of apoptosis in cells and pro-pregnancy responses mediated by IL-2 and INF-γ
incubated with T. cruzi antigens, indicating an anti-parasite respectively. The production of IL-17 is elevated in T. cruzi
pattern. During a normal pregnancy, the lymphocyte apop- infection and accentuated after zinc supplementation. IL-17
tosis is elevated compared to non-pregnancy models. The is a pro-inflammatory cytokine which induces the production

13
Acta Parasitologica

of IL-6, TNF-α, GM-CSF, and induces neutrophil migration 2. Bartmann C, Segerer SE, Rieger L, Kapp M, Sutterlin M, Kam-
[23, 55]. The cytokine controls T. cruzi infection by trapping merer U (2014) Quantification of the predominant immune cell
populations in decidua throughout human pregnancy. Am J
parasites in the endolysosomal compartment [19] and IL-17 Reprod Immunol 71:109–119. https​://doi.org/10.1111/aji.12185​
KO mice are highly susceptible to acute Chagas disease [30]. 3. Batista BL, Rodrigues JL, Nunes JA, Souza VC, Barbosa F Jr
However, an elevated proportion of IL-17 secreting cells (2009) Exploiting dynamic reaction cell inductively coupled
have been correlated with complicated pregnancies char- plasma mass spectrometry (DRC-ICP-MS) for sequential deter-
mination of trace elements in blood using a dilute-and-shoot pro-
acterized by miscarriage, preterm birth, and preeclampsia cedure. Anal Chim Acta 639:13–18. https​://doi.org/10.1016/j.
[15]. Thus, novel assays are required to elucidate the role aca.2009.03.016
of IL-17 in a model combining the period of conversion of 4. Benevides L, Milanezi CM, Yamauchi LM, Benjamim CF, Silva
acute to the chronic phase of Chagas disease and pregnancy. JS, Silva NM (2008) CCR2 receptor is essential to activate micro-
bicidal mechanisms to control Toxoplasma gondii infection in the
Corticosterone, an important stress hormone [27], is sensi- central nervous system. Am J Pathol 173:741–751. https​://doi.
tive to T. cruzi infection and/or zinc supplementation. High org/10.2353/ajpat​h.2008.08012​9
levels of corticosterone induce immune suppression [44] and 5. Bryan NS, Grisham MB (2007) Methods to detect nitric oxide
improve the rat susceptibility to T. cruzi [35]. In pregnancy, and its metabolites in biological samples. Free Radic Biol Med
43:645–657. https:​ //doi.org/10.1016/j.freera​ dbiom
​ ed.2007.04.026
the elevation of corticosterone levels limits the fetal growth, 6. Cardillo F, Voltarelli JC, Reed SG, Silva JS (1996) Regulation of
reducing the amino acid supply and density of blood vessels Trypanosoma cruzi infection in mice by gamma interferon and
in the placenta [51]. Thus, zinc demonstrates an anti-T. cruzi interleukin 10: role of NK cells. Infect Immun 64:128–134
and pro-pregnancy patterns, maintaining low levels of corti- 7. Carvalho CM, Silverio JC, da Silva AA, Pereira IR, Coelho JM,
Britto CC, Moreira OC, Marchevsky RS, Xavier SS, Gazzinelli
costerone in pregnant rats infected with T. cruzi. RT, da Gloria B-A, Lannes-Vieira J (2012) Inducible nitric oxide
This study reveals several immune aspects of zinc supple- synthase in heart tissue and nitric oxide in serum of Trypano-
mentation in pregnant Wistar rats in an experimental model soma cruzi-infected rhesus monkeys: association with heart
of the Chagas disease. Our results indicate that the immune injury. PLoS Negl Trop Dis 6:e1644. https:​ //doi.org/10.1371/journ​
al.pntd.00016​44
response induced by T. cruzi during pregnancy has a distinct 8. Chakraborty D, Rumi MA, Konno T, Soares MJ (2011) Natural
profile compared to non-pregnant models. Zinc supplemen- killer cells direct hemochorial placentation by regulating hypoxia-
tation promoted an immune response with potential for the inducible factor dependent trophoblast lineage decisions. Proc
T. cruzi control in parallel to a pro-gestation response. Thus, Natl Acad Sci USA 108:16295–16300. https​://doi.org/10.1073/
pnas.11094​78108​
the element contributes to the immune regulation, which is 9. Chasapis CT, Loutsidou AC, Spiliopoulou CA, Stefanidou ME
fundamental to the special condition of pregnancy. However, (2012) Zinc and human health: an update. Arch Toxicol 86:521–
new assays are necessary to describe the mechanisms of zinc 534. https​://doi.org/10.1007/s0020​4-011-0775-1
immune regulation during pregnancy, guiding a consistent 10. Chen L, Flies DB (2013) Molecular mechanisms of T cell co-
stimulation and co-inhibition. Nat Rev Immunol 13:227–242.
strategy for food supplementation in patients affected by the https​://doi.org/10.1038/nri34​05
Chagas disease. 11. Correa VR, Barbosa FG, Melo Junior CA, D’Albuquerque e Cas-
tro LF, Andrade Junior HF, Nascimento N (2014) Uneventful ben-
Acknowledgements  We would like to thank FAPESP for the fellow- znidazole treatment of acute Chagas disease during pregnancy: a
ship to C.M.B. da Costa (Grant number: 2013/04205-6). We also thank case report. Rev Soc Bras Med Trop 47:397–400
Fabiana Rosetto Morais and Vanessa Cristina de Oliveira Souza for the 12. da Costa CM, Brazao V, Collins Kuehn C, Rodrigues Oliveira
flow cytometry and HPLC assistance, respectively. LG, do Prado Junior JC, Sala MA, Abrahao AAC (2013) Zinc
and pregnancy: Marked changes on the immune response fol-
Author Contributions  CMBDC, JCDPJ, and AACA designed the lowing zinc therapy for pregnant females challenged with Trypa-
experiments, wrote, and revised the manuscript. CMBDC, MVF, FS, nosoma cruzi. Clin Nutr 32:592–598. https​://doi.org/10.1016/j.
and IFLG carried out the experimental work. LMP and FBJ analyzed clnu.2012.10.012
the data, wrote, and revised the manuscript. All authors read and 13. da Costa CMB, Pereira LM, Barbosa Jr F, do Prado JC, Abrahão
approved the final manuscript. AAC (2017) Chagas disease control and role of zinc supple-
mentation in pregnancy. Matters. https​://doi.org/10.19185​/matte​
rs.20161​20000​08
Compliance with Ethical Standards  14. da Costa CMB, Del Vecchio FM, Santello FH, Pereira LM, Toldo
MPA, do Prado Junior JC, Abrahao AAC (2018) Is the adaptive
Conflict of interest  The authors declare that they have no conflict of immune response in murine Trypanosoma cruzi infection influ-
interest. enced by zinc supplementation? Eur J Pharm Sci 111:330–336.
https​://doi.org/10.1016/j.ejps.2017.10.014
15. Darmochwal-Kolarz D, Michalak M, Kolarz B, Przegalinska-
Kalamucka M, Bojarska-Junak A, Sliwa D, Oleszczuk J (2017)
8. References The role of interleukin-17, interleukin-23, and transform-
ing growth factor-beta in pregnancy complicated by placen-
1. Abrahams VM, Straszewski-Chavez SL, Guller S, Mor G (2004) tal insufficiency. Biomed Res Int 2017:6904325. https​://doi.
First trimester trophoblast cells secrete Fas ligand which induces org/10.1155/2017/69043​25
immune cell apoptosis. Mol Hum Reprod 10:55–63. https​://doi. 16. Decote-Ricardo D, Nunes MP, Morrot A, Freire-de-Lima
org/10.1093/moleh​r/gah00​6 CG (2017) Implication of apoptosis for the pathogenesis of

13
Acta Parasitologica

Trypanosoma cruzi infection. Front Immunol 8:518. https​: // 31. Mor G, Cardenas I (2010) The immune system in pregnancy: a
doi.org/10.3389/fimmu​.2017.00518​ unique complexity. Am J Reprod Immunol 63:425–433. https​
17. Duthie MS, Kahn SJ (2005) NK cell activation and protection ://doi.org/10.1111/j.1600-0897.2010.00836​.x
occur independently of natural killer T cells during Trypano- 32. Oliveira LRC, Picka MCM, Nicolete VC, Calvi SA, Marcondes-
soma cruzi infection. Int Immunol 17:607–613. https​: //doi. Machado J (2012) Organ tropism during the acute and chronic
org/10.1093/intim​m/dxh23​9 phases of Trypanosoma cruzi infection in BALB/c mice. J
18. Erdmann H, Rossnagel C, Bohme J, Iwakura Y, Jacobs T, Venom Anim Toxins incl Trop Dis 18:34–43. https​: //doi.
Schaible UE, Holscher C (2013) IL-17A promotes macrophage org/10.1590/S1678​-91992​01200​01000​05
effector mechanisms against Trypanosoma cruzi by trapping 33. Van Overtvelt L, Andrieu M, Verhasselt V, Connan F, Chop-
parasites in the endolysosomal compartment. Immunobiology pin J, Vercruysse V, Goldman M, Hosmalin A, Vray B (2002)
218:910–923. https​://doi.org/10.1016/j.imbio​.2012.10.005 Trypanosoma cruzi down-regulates lipopolysaccharide-induced
19. Erdmann H, Behrends J, Holscher C (2016) During acute MHC class I on human dendritic cells and impairs antigen
experimental infection with the reticulotropic Trypanosoma presentation to specific CD8(+) T lymphocytes. Int Immunol
cruzi strain Tulahuen IL-22 is induced IL-23-dependently but 14:1135–1144
is dispensable for protection. Sci Rep 6:32927. https​: //doi. 34. Paiva CN, Figueiredo RT, Kroll-Palhares K, Silva AA, Silverio
org/10.1038/srep3​2927 JC, Gibaldi D, Pyrrho Ados S, Benjamim CF, Lannes-Vieira J,
20. Fraccaroli L, Alfieri J, Larocca L, Calafat M, Mor G, Leirós CP, Bozza MT (2009) CCL2/MCP-1 controls parasite burden, cell
Ramhorst R (2009) A potential tolerogenic immune mechanism infiltration, and mononuclear activation during acute Trypano-
in a trophoblast cell line through the activation of chemokine- soma cruzi infection. J Leukoc Biol 86:1239–1246. https​://doi.
induced T cell death and regulatory T cell modulation. Hum org/10.1189/jlb.03091​87
Reprod 24:166–175. https​://doi.org/10.1093/humre​p/den34​4 35. Pereira M, Soares C, Canuto GA, Tavares MF, Colli W, Alves
21. Gil-Jaramillo N, Motta FN, Favali CB, Bastos IM, Santana MJ (2015) Down regulation of NO signaling in Trypanosoma
JM (2016) Dendritic cells: a double-edged sword in immune cruzi upon parasite-extracellular matrix interaction: changes
responses during Chagas disease. Front Microbiol 7:1076. https​ in protein modification by nitrosylation and nitration. PLoS
://doi.org/10.3389/fmicb​.2016.01076​ Negl Trop Dis 9:e0003683. https​: //doi.org/10.1371/journ​
22. Haase H, Rink L (2014) Multiple impacts of zinc on immune al.pntd.00036​83
function. Metallomics 6:1175–1180. https​://doi.org/10.1039/ 36. Perez AR, Bertoya AA, Revelli S, Garcia F (2011) A high cor-
c3mt0​0353a​ ticosterone/DHEA-s ratio in young rats infected with Trypano-
23. Jovanovic DV, Di Battista JA, Martel-Pelletier J, Jolicoeur FC, soma cruzi is associated with increased susceptibility. Mem Inst
He Y, Zhang M, Mineau F, Pelletier JP (1998) IL-17 stimulates Oswaldo Cruz 106:416–423
the production and expression of proinflammatory cytokines, 37. Prasad AS (1995) Zinc: an overview. Nutrition 11:93–99
IL-beta and TNF-alpha, by human macrophages. J Immunol 38. Robben PM, LaRegina M, Kuziel WA, Sibley LD (2005) Recruit-
160:3513–3521 ment of Gr-1+ monocytes is essential for control of acute toxo-
24. Kaminski VL, Ellwanger JH, Chies JAB (2019) Extracellular plasmosis. J Exp Med 201:1761–1769. https​://doi.org/10.1084/
vesicles in host-pathogen interactions and immune regulation— jem.20050​054
exosomes as emerging actors in the immunological theater of 39. Rottenberg M, Cardoni RL, Andersson R, Segura EL, Orn A
pregnancy. Heliyon 5:e02355. https​://doi.org/10.1016/j.heliy​ (1988) Role of T helper/inducer cells as well as natural killer cells
on.2019.e0235​5 in resistance to Trypanosoma cruzi infection. Scand J Immunol
25. Keen CL, Clegg MS, Hanna LA, Lanoue L, Rogers JM, Das- 28:573–582
ton GP, Oteiza P, Uriu-Adams JY (1605s) The plausibility 40. Sales Junior PA, Molina I, Fonseca Murta SM, Sanchez-Montalva
of micronutrient deficiencies being a significant contributing A, Salvador F, Correa-Oliveira R, Carneiro CM (2017) Experi-
factor to the occurrence of pregnancy complications. J Nutr mental and clinical treatment of chagas disease: a review. Am
133:1597s–1605s. https​://doi.org/10.1093/jn/133.5.1597S​ J Trop Med Hyg 97:1289–1303. https​://doi.org/10.4269/ajtmh​
26. Kuziel WA, Morgan SJ, Dawson TC, Griffin S, Smithies O, .16-0761
Ley K, Maeda N (1997) Severe reduction in leukocyte adhesion 41. Schroder K, Hertzog PJ, Ravasi T, Hume DA (2004) Interferon-
and monocyte extravasation in mice deficient in CC chemokine gamma: an overview of signals, mechanisms and functions. J
receptor 2. Proc Natl Acad Sci USA 94:12053–12058 Leukoc Biol 75:163–189. https​://doi.org/10.1189/jlb.06032​52
27. Liu W, Yuen EY, Yan Z (2010) The stress hormone corticoster- 42. Serbina NV, Jia T, Hohl TM, Pamer EG (2008) Monocyte-medi-
one increases synaptic alpha-amino-3-hydroxy-5-methyl-4-isox- ated defense against microbial pathogens. Annu Rev Immunol
azolepropionic acid (AMPA) receptors via serum- and gluco- 26:421–452. https​://doi.org/10.1146/annur​ev.immun​ol.26.02160​
corticoid-inducible kinase (SGK) regulation of the GDI-Rab4 7.09032​6
complex. J Biol Chem 285:6101–6108. https​://doi.org/10.1074/ 43. Silva LHP, Nussenzweig V (1953) Sobre uma cepa de Trypano-
jbc.M109.05022​9 soma cruzi altamente virulenta para o camundongo branco. Folia
28. Marcipar IS, Risso MG, Silber AM, Revelli S, Marcipar AJ clinica et biologica 20:191–207
(2001) Antibody maturation in Trypanosoma cruzi-infected rats. 44. Sisti G, Kanninen TT, Witkin SS (2016) Maternal immunity
Clin Diagn Lab Immunol 8(4):802–805. https​://doi.org/10.1128/ and pregnancy outcome: focus on preconception and autophagy.
CDLI.8.4.802-805.2001 Genes Immun 17:1–7. https​://doi.org/10.1038/gene.2015.57
29. Miyahira Y, Katae M, Kobayashi S, Takeuchi T, Fukuchi Y, Abe 45. Sundareswaran L, Srinivasan S, Wankhar W, Sheeladevi R (2017)
R, Okumura K, Yagita H, Aoki T (2003) Critical contribution of Effect of Scoparia dulcis on noise stress induced adaptive immu-
CD28-CD80/CD86 costimulatory pathway to protection from nity and cytokine response in immunized Wistar rats. J Ayurv
Trypanosoma cruzi infection. Infect Immun 71:3131–3137 Integr Med 8:13–19. https​://doi.org/10.1016/j.jaim.2016.10.004
30. Miyazaki Y, Hamano S, Wang S, Shimanoe Y, Iwakura Y, 46. Sykes L, MacIntyre DA, Yap XJ, Ponnampalam S, Teoh TG, Ben-
Yoshida H (2010) IL-17 is necessary for host protection against nett PR (2012) Changes in the Th1:Th2 cytokine bias in preg-
acute-phase Trypanosoma cruzi infection. J Immunol 185:1150– nancy and the effects of the anti-inflammatory cyclopentenone
1157. https​://doi.org/10.4049/jimmu​nol.09000​47 prostaglandin 15-deoxy-Delta(12,14)-prostaglandin J2. Mediat
Inflamm 2012:416739. https​://doi.org/10.1155/2012/41673​9

13
Acta Parasitologica

47. Teixeira MM, Gazzinelli RT, Silva JS (2002) Chemokines, mice. J Physiol 590:5529–5540. https​://doi.org/10.1113/jphys​
inflammation and Trypanosoma cruzi infection. Trends Parasitol iol.2012.23942​6
18:262–265 52. Vespa GN, Cunha FQ, Silva JS (1994) Nitric oxide is involved in
48. Tezabwala BU, Johnson PM, Rees RC (1989) Inhibition of preg- control of Trypanosoma cruzi-induced parasitemia and directly
nancy viability in mice following IL-2 administration. Immunol- kills the parasite in vitro. Infect Immun 62:5177–5182
ogy 67:115–119 53. World Health Organization (2018) Chagas disease (American
49. Totemeyer S, Sheppard M, Lloyd A, Roper D, Dowson C, trypanosomiasis). Retrieved from World Health Organization
Underhill D, Murray P, Maskell D, Bryant C (2006) IFN-gamma website. https​://www.who.int/media​centr​e/facts​heets​/fs340​/en/.
enhances production of nitric oxide from macrophages via a Accessed 14 May 2018
mechanism that depends on nucleotide oligomerization domain-2. 54. Xu YY, Wang SC, Li DJ, Du MR (2017) Co-signaling molecules
J Immunol 176:4804–4810 in maternal-fetal immunity. Trends Mol Med 23:46–58. https​://
50. Vallee BL, Falchuk KH (1993) The biochemical basis of zinc doi.org/10.1016/j.molme​d.2016.11.001
physiology. Physiol Rev 73:79–118. https:​ //doi.org/10.1152/physr​ 55. Yao Z, Painter SL, Fanslow WC, Ulrich D, Macduff BM, Spriggs
ev.1993.73.1.79 MK, Armitage RJ (1995) Human IL-17: a novel cytokine derived
51. Vaughan OR, Sferruzzi-Perri AN, Fowden AL (2012) Maternal from T cells. J Immunol 155:5483–5486
corticosterone regulates nutrient allocation to fetal growth in

13

You might also like