ArticlePDF Available

Molecular identification of Trypanosoma cruzi in domestic animals in municipalities of the State of Rio Grande do Norte, Brazil

Authors:

Abstract and Figures

Trypanosoma cruzi, the etiologic agent of American trypanosomiasis, is a vector-borne zoonotic parasite which has been little studied regarding its infection in domestic animals. In this study, we evaluated the occurrence of natural infection by T. cruzi in farm animals using molecular markers and phylogenetic analysis in blood clot samples of 60 sheep (Ovis aires), 22 goats (Capra hircus), and 14 horses (Equus caballus) in eight municipalities located in an infection risk area in the state of Rio Grande do Norte (RN), Northeast Region of Brazil. Trypanosoma spp. infection was identified by amplifying the rRNA 18S SSU gene in 48.9% of the samples. The SH022 sample showed 99.8% similarity with the Y strain of T. cruzi in phylogeny, grouped in the DTU II clade. Blood clots of sheep, goats, and horses detected T. cruzi kDNA in 28.3% (17/60), 22.7% (5/22), and 15.4% (2/14) of the samples, respectively. These animals were distributed in the three studied mesoregions throughout the state of RN. The identification of natural infection in domestic animals contributes to expand the epidemiological transmission scenario in an area where T. brasiliensis is the main vector.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Parasitology Research
https://doi.org/10.1007/s00436-022-07719-w
RESEARCH
Molecular identification ofTrypanosoma cruzi indomestic animals
inmunicipalities oftheState ofRio Grande doNorte, Brazil
VicenteToscanodeAraújo‑Neto1· AndressaNoronhaBarbosa‑Silva1· NathanRaviMedeirosHonorato2·
LetíciaMikardyaLimaSales3· RenatadeCassiaPires1· CarlosRamondoNascimentoBrito4·
PauloMarcosdaMattaGuedes4· LúciaMariadaCunhaGalvão1,2· AntoniaClaudiaJácomedaCâmara1,4
Received: 3 August 2022 / Accepted: 5 November 2022
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2022
Abstract
Trypanosoma cruzi, the etiologic agent of American trypanosomiasis, is a vector-borne zoonotic parasite which has been
little studied regarding its infection in domestic animals. In this study, we evaluated the occurrence of natural infection by
T. cruzi in farm animals using molecular markers and phylogenetic analysis in blood clot samples of 60 sheep (Ovis aires),
22 goats (Capra hircus), and 14 horses (Equus caballus) in eight municipalities located in an infection risk area in the state
of Rio Grande do Norte (RN), Northeast Region of Brazil. Trypanosoma spp. infection was identified by amplifying the
rRNA 18S SSU gene in 48.9% of the samples. The SH022 sample showed 99.8% similarity with the Y strain of T. cruzi in
phylogeny, grouped in the DTU II clade. Blood clots of sheep, goats, and horses detected T. cruzi kDNA in 28.3% (17/60),
22.7% (5/22), and 15.4% (2/14) of the samples, respectively. These animals were distributed in the three studied mesoregions
throughout the state of RN. The identification of natural infection in domestic animals contributes to expand the epidemio-
logical transmission scenario in an area where T. brasiliensis is the main vector.
Keywords Trypanosoma cruzi· Natural infection· Phylogenetic analysis· American trypanosomiasis
Introduction
Chagas disease (CD) or American trypanosomiasis, caused
by Trypanosoma cruzi Chagas, 1909, affects about 6 to 7
million people in the world and another 75 million lives in
areas at risk for transmission (WHO 2022). CD presents
high morbidity and mortality in endemic countries, with
approximately 12,000 deaths per year, and is considered
a public health problem in Latin America (PAHO 2022).
This continent presents a high density of insects of the Tri-
atominae subfamily (Hemiptera, Reduviidae), which are
Section Editor: Vyacheslav Yurchenko
* Antonia Claudia Jácome da Câmara
acjcamara@ufrnet.br
Vicente Toscano de Araújo-Neto
toscanorn1@gmail.com
Andressa Noronha Barbosa-Silva
noronha.andressa@gmail.com
Nathan Ravi Medeiros Honorato
nathanhonorato@hotmail.com
Letícia Mikardya Lima Sales
lmlsaless@gmail.com
Renata de Cassia Pires
renatacapires@gmail.com
Carlos Ramon do Nascimento Brito
crnbrito@yahoo.com.br
Paulo Marcos da Matta Guedes
guedespmm@gmail.com
Lúcia Maria da Cunha Galvão
luciabhster@gmail.com
1 Graduate Program inPharmaceutical Sciences, Federal
University ofRio Grande Do Norte, Natal59012-570, Brazil
2 Graduate Program inParasitology, Federal University
ofMinas Gerais, Belo Horizonte, BeloHorizonte31270-901,
Brazil
3 Undergraduate Course inPharmacy, Federal University
ofRio Grande Do Norte, Natal59012-570, Brazil
4 Graduate Program inParasite Biology, Federal University
ofRio Grande Do Norte, Natal59064-741, Brazil
Parasitology Research
1 3
vectors of T. cruzi (Chagas 1909; Lent and Wygodzinsky
1979; Lima-Neiva etal. 2021).
Trypanosoma cruzi is a digenetic protozoan which
infects several animal orders, for example, Didelphi-
morphia, Carnivora, Rodentia, Chiroptera, and Primate,
including human beings, demonstrating the generalist and
successful character of this parasite (Jansen etal. 2018).
Transmission to vertebrate animals can happen in a classi-
cal way by contamination in the vector’s feces, or orally by
predation of infected reservoirs by susceptible ones. The
interaction dynamic between the host and parasite is com-
plex, and every animal has a unique role in the transmis-
sion cycle in different areas (Jansen and Roque 2010). The
infection prevalence of an animal, host capacity to infect
vectors, and the host–triatomine contact are factors which
determine the importance of a species as a pathogen res-
ervoir transmitted by a vector (Cohen and Gurtler 2001).
From an ecological point of view, CD is typical in rural
areas where the triatomine interacts with domestic and
sylvatic animals, transmitting the parasite (Oliveira-Lima
etal. 2000). This interaction is quite evident in peridomi-
cile environments, as they are important epidemiological
environments which act as a link between anthropized and
wild zones. Peridomicile transmission involves domestic
reservoirs and sylvatic triatomines attracted to the dwell-
ing by light searching for feeding sources, or well-adapted
vectors to human habitations which can establish their col-
onies in this environment (Dias etal. 2000). Synanthropic
vertebrates, like opossums, bats, and rodents also play an
important role in the transmission of T. cruzi to humans,
connecting both environments in a single epidemiological
cycle (Fernandes etal. 1989; Yefi-Quinteros etal. 2018;
Drozino etal. 2019).
Infestation and colonization of Triatoma brasiliensis
Neiva, 1911, the main vector in the state of Rio Grande do
Norte (RN), Northeast Region of Brazil, has been reported
in recent studies (Barbosa-Silva etal. 2019; Araújo-Neto
etal. 2019; Honorato etal. 2021; Lima-Neiva etal. 2021).
This species is frequently found in ecotopes like corrals,
brick/tile piles, and chicken coops (Barbosa-Silva etal.
2016; Lilioso etal. 2020), associated to domestic animals
such as sheep (Ovis aires), goats (Capra hircus), and
chickens (Gallus gallus), and synanthropic animals like
Galea spixii (Barbosa-Silva 2017; Bezerra etal. 2018;
Lima-Neiva etal. 2021). Moreover, high infection rates
in dogs (Canis familiaris) have also been demonstrated in
the same area (Araújo-Neto etal. 2019). However, the role
of domestic animals in establishing and maintaining the
transmission cycle of T. cruzi has not yet been completely
elucidated. In this context, we evaluated the natural infec-
tion occurrence of T. cruzi by molecular analysis in sheep,
goats, and horses in municipalities of RN State, Brazil.
Materials andmethods
Study area andpopulation
The state of RN is located in the Northeast Region of
Brazil and has an area of 52,809.6 km2, divided into 167
municipalities distributed in four mesoregions: West,
Central, Agreste, and East. A semi-arid, arid, or dry sub-
humid climate predominates in the state, ranging from
the Agreste mesoregion to the northern coast (IDEMA
2020). Livestock production mostly comprises small herds
of goats, sheep, and horses of resistant and well-adapted
breeds mainly used as food support (IBGE 2021).
The surveyed areas were selected according to the fol-
lowing criteria: (i) medium or high risk of vectoral trans-
mission of T. cruzi (Barbosa-Silva etal. 2019); (ii) infec-
tion occurrence in human, dogs, or sylvatic animals (Brito
etal. 2012; Martins etal. 2015; Araújo-Neto etal. 2019);
and (iii) occurrence of infected triatomines (Barbosa-Silva
etal. 2016, 2019; Vargas etal. 2018; Liliosoet al. 2020;
Honorato etal. 2021). This study was conducted in 17
rural communities in the following municipalities: João
Câmara in the Agreste mesoregion, Alexandria, Marcelino
Vieira, Upanema, Lajes, Caraúbas and Campo Grande in
the West mesoregion, and Afonso Bezerra in the Central
mesoregion (Table1).
Sampling andDNA extraction
Blood was initially collected from 60 sheep, 22 goats, and
14 horses and submitted to the microhematocrit method to
search for motile parasites (Woo 1970). A blood clot was
obtained from all samples and individually stored in abso-
lute ethanol until the moment of DNA extraction (Rodri-
gues etal. 2019). DNA extraction following the protocol
of Garcia etal (2018).
Nested PCR ofrRNA 18S SSU gene
The 18S SSUrRNA gene was detected as proposed by
Noyes etal. (1999) using two pairs of primers. In the first
step, the TRY927F (5-GAA ACA AGA AAC ACG GGA
G-3) and TRY927R (5-CTA CTG GGC AGC TTGGA-3)
primers were used, and then, the SSU561F (5-TGG GAT
AAC AAA GGA GCA -3) and SSU561R primers (5-CTG
AGA CTG TAA CCT CAA AGC-3) were used in the sec-
ond step (Smith etal. 2008). The amplicons between 600
and 1000bp were visualized by electrophoresis in a 6%
polyacrylamide gel stained with silver salts (Santos etal.
1993). In addition, RN28 (Câmara etal. 2013) and 3188
Parasitology Research
1 3
(Martins etal. 2015) strains of T. cruzi were used as posi-
tive controls. The samples had their final DNA concentra-
tion adjusted from 5 to 20ng/μL.
Purification, sequencing, andphylogenetic analysis
Purification was performed using the Exo SAP-IT™ PCR
Product Clean up (Thermo Fisher Scientific, Waltham, USA)
following the manufacturer’s protocol. Sequencing reactions
were carried out using the BigDye® Terminator v 3.1 Cycle
Sequencing Kit (Applied Biosystems, Foster City, USA).
The sequencing reactions were performed in an ABI 3500
DNA Analyzer (Thermo Fisher Scientific, USA).
Electropherograms were analyzed, and consensus
sequences were generated using the Mega X software pro-
gram (Kumar etal. 2018). The consensus sequences for
phylogenetic analysis were compared to samples depos-
ited in the NCBI/GenBank database (National Center for
Table 1 Distribution of goats,
sheep, and equine infected by
Trypanosoma cruzi identified
with kDNA and 18S SSU
in rural communities of the
mesoregions and municipalities
in the State of Rio Grande do
Norte
Mesoregions/Municipality Hosts species
Capra hircus Ovis aries Equus caballus
NkDNA 18S NkDNA 18S NkDNA 18S
Agreste
João Câmara 3 3 3 15 11 11 1 1 1
Central
Afonso Bezerra 2 2 2 - - - - - -
West
Marcelino Vieira 6 0 2 15 3 5 - - -
Alexandria 3 0 0 17 2 9 12 1 0
Campo Grande 7 0 6 2 0 2 - - -
Caraúbas - - - 6 0 5 - - -
Lajes 1 0 1 4 1 1 1 0 0
Upanema - - - 1 0 1 - - -
Total (%) 22 5 (22.7) 14 (63.6) 60 17 (28.3) 34 (56.6) 14 2 (35.4) 1 (7.7)
Table 2 Sample, molecular identification, discrete typing unit (DTU), hosts species, GenBank code, and reference of samples used in genetic
sequencing analysis
DTU discrete typing unit by molecular identification of gene 18S SSU; *ingroup; #outgroup
Sample ID Molecular identification DTU Host species GenBank code Reference
bCth 743*T. cruzi TcI Cerdocyon thous MH411618 Rodrigues etal. 2019
bCth 744*T. cruzi TcI Cerdocyon thous MH411619 Rodrigues etal. 2019
CPAPGM 855*T. cruzi TcI Cerdocyon thous MH411626 Rodrigues etal. 2019
APAGM 850*T. cruzi TcI Cerdocyon thous MH411625 Rodrigues etal. 2019
CPAGM 796*T. cruzi TcI Cerdocyon thous MH411623 Rodrigues etal. 2019
CPAGM 848*T. cruzi TcI Cerdocyon thous MH411624 Rodrigues etal. 2019
Colombiana*T. cruzi TcI Homo sapiens AF239980 Kawashita etal., 2001
LBT 6867*T. cruzi TcII Artibeus lituratus MH411627 Rodrigues etal. 2019
YL 881*T. cruzi TcII Marmosopsincanus MH558662 Rodrigues etal. 2019
Y*T. cruzi TcII Homo sapiens AF301912 Kawashita etal., 2001
LBCE 19,688*T. cruzi TcII Didelphis marsupialis MH411637 Rodrigues etal. 2019
LBCE 19,675*T. cruzi TcII Didelphis marsupialis MH411635 Rodrigues etal. 2019
PCE 383*T. cruzi TcII Marmosa sp. MH411630 Rodrigues etal. 2019
EM 897*T. cruzi TcII Artibeus fimbriatus MH411629 Rodrigues etal. 2019
EM 712*T. cruzi TcIII Artibeus lituratus MH558661 Rodrigues etal. 2019
EM 693*T. cruzi TcIII Carollia perspicillata MH411642 Rodrigues etal. 2019
RN02*T. cruzi TcIII Triatoma brasiliensis OP132415 Câmara etal. 2010
SH022 T. cruzi TcII Ovis aries OP132414 Araujo-Neto etal. in
press
PCE 51# T. cascavelli Didelphis albiventris MH411650 Rodrigues etal. 2019
Parasitology Research
1 3
Biotecnology Information https:// www. ncbi. nlm. nih. gov/
BLAST/) using the BLAST tool, program selection optimize
for highly similar sequences (megablast). Table2 shows the
sequences of ingroups and outgroups. These sequences
were submitted to multiple alignments with the MAFFT v.
7 online program (Katoh and Standley 2013). The genome
alignments were inspected and corrected on Mega X; then,
the most suitable evolutionary model was tested using the
Model Finder plugin on the PhyloSuite v. 1.2.2 (Zhang etal.
2020). Next, phylogenetic analysis was performed using two
maximum likelihood models: (I) MrBayes v. 3.2.6 (Ronquist
etal. 2012) was used for phylogenetic analysis by Bayesian
inference with GTR + F + I model, two parallel runs, and
10,000 bootstrap replicates. About 25% of the initial data
sampled were discarded as burn-in; (II) IQ-TREE (Nguyen
etal. 2015) using the TN + I model for 10,000 standard
bootstraps (Minh etal. 2013), as well as the Shimodaira-
Hasegawa approximate likelihood-ratio test (Guindon etal.
2010). A phylogenetic tree was generated, evaluated, and
interpreted using Figtree v. 1.4.4 and edited in Inkscape.
T. cruzi kDNA detection
PCR for detecting T. cruzi kDNA was performed as pre-
viously described (Gomes etal. 1998), targeting constant
regions of minicircles using primers described by Vallejo
etal (1999). Amplicons of 330bp were visualized in poly-
acrylamide gels as previously described. The SM76 strain
(Martins etal. 2015) of T. cruzi was used as positive control
and non-infected human blood as negative control.
Results
Trypanosoma spp. infection
Motile parasites were not detected in the samples by the
microhematocrit technique. Table1 shows a wide distri-
bution of 18S-positive samples throughout RN State. All
animal species in the municipality of João Câmara were
Fig. 1 a Representative polyacrylamide gel electrophoresis show-
ing the amplification of rRNA 18S SSU gene of Trypanosoma spp
stained by silver from blood clot samples from goats (GOA), sheep
(SH), and controls. Fragments between 600 and 1000bp were con-
sidered positive. M: Molecular size marked. Lines 2–7: Trypanosoma
spp. samples SH056, SH057, SH058, GOA012, GOA013, GOA014.
Line 8, 10 and 11: T. cruzi strain-positive controls RN2, 3188. Line 9,
SH004-negative sample, NC. Line 12, NC: negative control. b Rep-
resentative polyacrylamide gel electrophoresis showing the ampli-
fication of kDNA of T. cruzi from blood clot samples from goats
(GOA) and sheep (SH). Fragment of 330bp was considered positive.
M Molecular size marked. Line 2: T. cruzi strain-positive controls:
SM76. Lines 3–10: T. cruzi-positive samples: GOA003. GOA004,
SH008, SH009, SH010, SH011, SH012, SH013. NC: negative control
Parasitology Research
1 3
infected, while only sheep and goats were found positive in
the municipalities of Marcelino Vieira, Campo Grande, and
Lajes. On the other hand, only goats were infected in Afonso
Bezerra, while only sheep were infected in Alexandria,
Caraúbas, and Upanema. In total, Trypanosoma spp. DNA
was detected in more than 50% of the sheep clot samples.
rRNA 18S SSU gene amplified fragments between 600
and 1000bp in 48.9% (48/96) of samples collected in all
investigated areas. A representative polyacrylamide gel of
electrophoresis to show the amplicon pattern produced in
this reaction can be seen in Fig.1a.
Trypanosoma cruzi identification
From 48 positive samples for 18S amplification, only
8.3% (4) were submitted to DNA sequencing procedures,
and only one (ovine sample SH022) presented an electro-
pherogram with good quality. This sample was collected
in João Câmara, and its consensus sequence was submit-
ted to BLASTn tool, which matched 99.8% identity with
DTU II Y strain (AF301912) of T. cruzi. The Bayesian
phylogenetic tree grouped SH022 in DTU II clade was also
associated with Y and other strains of this genotype with a
bootstrap value higher than 0.98 (Fig.2). The RN02 sam-
ple, genotyped by Câmara etal. (2010) as TcIII from a T.
brasiliensis specimen captured in RN State, was grouped
with the sequences from GenBank forming a DTU III clade.
Sequences from DTU I were grouped all together in a father
branch. The outgroup, represented by Trypanosoma cas-
cavelli Pessoa and De Biasi, 1972 (MH411650), formed an
external clade (Fig.2).
T. cruzi kDNA detection
PCR detected T. cruzi kDNA in 28.3% (17/60), 22.7%
(5/22), and 15.4% (2/14) of samples of sheep, goats, and
horses, respectively, demonstrating a high infection rate. A
representative polyacrylamide gel of electrophoresis to show
the amplicon pattern of 330bp produced in this reaction
can be seen in Fig.1b. All samples of horses and goats and
73.3% (11/15) of sheep were infected in João Câmara. All
municipalities presented at least one positive animal species,
except for Campo Grande (Table1, Fig.3).
Discussion
The occurrence of natural infection and phylogenetic analy-
sis of T. cruzi was reported in sheep, goats, and horses in
municipalities of medium and high risk of vectorial trans-
mission of this parasite in the state of RN, Brazil. A broad
distribution of infected animals was observed, as confirmed
by T. cruzi kDNA identification and detection of DTU II by
sequencing and phylogeny of rRNA 18S SSU gene.
Fig. 2 Bayesian phylogenetic tree based on gene rRNA 18SSSU
using the sequences of SH022, RN02 isolated from T. brasiliensis
(CAMARA etal., 2010) and the sequence obtained from GenBank.
The numbers in branches correspond to support values, with 1.0
being the highest similarity value. The numbers in nodes are the sup-
port values ordered as Bayesian inference/IQ-TREE. T. cascavelli was
used as the outgroup
Parasitology Research
1 3
Infection of farm animals by T. cruzi is poorly reported
using molecular or serological techniques. Infection in
sheep was described in Argentina by amplification of sat-
ellite DNA (Sat-DNA) by real-time PCR (Wehrendt etal.
2019). A high percentage of infected goats was detected in
locations with hot and dry climates in the Northeast Region
of Brazil, constituting areas with similar conditions of the
present work (Muñoz-San Martín etal. 2020). In addition,
only horses in Venezuela have been found positive by kDNA
and Sat-DNA (Herrera etal. 2022).
Studies regarding blood surveys have shown high infec-
tion rates in goats and sheep by the indirect hemagglutina-
tion test in Chile (Schenone etal. 1991). Goats in Northeast
Brazil were highly infected in Paraíba State (Fuentes-Cas-
tillo etal. 1988), while sheep and goats in Ceará State dem-
onstrated low reactivity percentages by indirect immunofluo-
rescence (Bezerra etal. 2014).
The role of these animals in spreading or maintaining
T. cruzi seems to be variable in different transmission
cycles. These animals are mostly part of a semi-intensive
livestock production system in the Northeast Region (i.e.,
they are free in the pasture during the day and collected
and put into corrals at night), increasing their chance of
exposure to the vectors. These ecotopes were already
reported sheltering triatomines in the same area sur-
veyed in the present study (Barbosa-Silva etal. 2016)
and in other areas of South America (Cecere etal. 1997;
Chartier and Crocco 2007; Gürtler etal. 2014; Carbajal-
de-la-Fuente etal. 2017). The DNA of these animals was
detected in vectors found in several ecotopes of peridom-
icile areas in Ceará (Valença-Barbosa etal. 2015) and in
Caraúbas (Barbosa-Silva, 2017) and Caicó (Lima-Neiva
etal. 2021), respectively located in the West and Cen-
tral mesoregions of RN State. Moreover, these animals
have also been found as a food source of triatomines
in sylvatic areas of RN (Barbosa-Silva, 2017). From an
anthropocentric point of view, these data suggest that
goats and sheep can act as a link between sylvatic and
peridomiciliar cycles (Ashford 1997), not only in RN
State but also in other endemic areas.
Fig. 3 Distribution of animal samples positive for T. cruzi infection
by kDNA detection in Rio Grande do Norte State. Municipalities sur-
veyed are highlighted in grey and represented by the following num-
bers: 1: João Câmara; 2: Lajes; 3: Afonso Bezerra; 4: Upanema; 5:
Campo Grande; 6: Caraúbas; 7: Alexandria; and 8: Marcelino Vieira
Parasitology Research
1 3
Herein, we have reported the natural infection of T. cruzi
in equines in Brazil for the first time. Ikenga and Richer-
son (1984) identified positive horses by serology in Texas,
USA. Molecular markers posteriorly identified infection in a
10-year-old quarter horse with neurological disturbances in
the same state, which also presented amastigote-like forms
in its spinal cord (Bryan etal. 2016). Taken together, our
findings and the literature data still cannot provide enough
information to suggest that horses have a role in the cycle of
T. cruzi. Thus, studies about parasite load and susceptibility
to blood meal of triatomines need to be performed.
Literature data in the last decade have also demonstrated
the diversity of T. cruzi populations which circulates through
anthropic and sylvatic cycles in RN State. Until this moment,
DTUs I, II, and III were found in humans and also in vectors
such as T. brasiliensis, Triatoma pseudomaculata Corrêa and
Espínola, 1964, and Panstrongylus lutzi Neiva and Pinto,
1923(Câmara etal. 2010; Martins etal. 2015; Honorato etal.
2021). Identification of DTU II in sheep adds a new variable
to understanding the interaction of animals and vectors in
the peridomicile areas in the surveyed region.
Amplification of 18S to identify Trypanosoma spp.
revealed some positive samples with T. cruzi. However, oth-
ers which were not identified can suggest the occurrence of
other trypanosomes like Trypanosoma vivax Ziemann, 1905,
and Trypanosoma evansi Evans, 1880. Both species have
been reported in South America (Guerra etal. 2008; Colpo
etal. 2005). The former is a protozoan with great economic
and veterinary importance which can cause abortion and
was identified in bovines in RN State (Batista etal. 2018).
The latter has been described in the Central-West Region of
Brazil (Costa etal. 2019), which is endemic for this infec-
tion, and in the states of Rio Grande do Sul (Rodrigues etal.
2005), Amazonas (Filgueiras etal. 2019), and Bahia (Costa
etal. 2019), but with no reports in RN State.
Conclusion
Identifying the high distribution of natural infection by T.
cruzi in farm animals contributes to amplify the epidemio-
logical transmission scenario in the areas where we sug-
gest that sheep and goats can be a link between sylvatic and
anthropic cycles. It is necessary to perform studies on horses
to deeper investigate the aspects about infection and its rel-
evance as a reservoir.
Acknowledgements The authors are grateful to the Secretariat of State
for Public Health, represented by the health authorities and health
agents of the Municipal Health Secretariats of Alexandria, Caraúbas,
Campo Grande, Marcelino Vieira, Upanema, Lajes, and João Câmara,
for their indispensable support in field activities and for the provision
of data during the development of this study.
Author contribution Vicente Toscano de Araújo-Neto, Andressa
Noronha Barbosa-Silva, Nathan Ravi de Medeiros Honorato, Antonia
Claudia Jácome da Câmara, and Renata de Cassia Pires designed the
experiments and wrote the original draft. Carlos Ramon do Nascimento
Brito and Letícia Micardya Lima Sales contributed to the study design.
Antonia Claudia Jácome da Câmara, Paulo Marcos da Matta Guedes,
and Lúcia Maria da Cunha Galvão conceptualized and reviewed the
manuscript. All authors have read the paper and approved the final
version.
Funding This study was supported by the Conselho Nacional de
Desenvolvimento Científico e Tecnológico (CNPq) (MCTI/CNPq/
Edital Universal 2016 grant no. 423966/2016–2). LMCG is a CNPq
fellow, and NRMH, ANBS, RCP, and LMLS thank the Coordenação
de Aperfeiçoamento de Pessoal de Nível Superior (CAPES) for grant-
ing scholarships.
Data availability The sequences were submitted to GenBank under the
following access numbers: OP132414–SH022 and OP132415–RN02.
The isolate RN02 in all blood samples are in the LABIOPAR collection
at Department of Clinical and Toxicological Analyses/UFRN.
Declarations
Ethical approval This study was approved by Ethics Committee on the
Use of Animals of Federal University of Rio Grande do Norte (CEUA-
UFRN), protocol no. 134.062/2018, Natal, RN.
Consent for publication All authors gave their consent for publication.
Conflict of interest The authors declare there are no conflicts of inter-
est.
References
Araújo-Neto VT, Honorato NRM, Oliveira-Santana R, Barbosa-Silva
AN, Guedes PMM, Chiari E, Galvão LM, Câmara ACJ (2019)
Trypanosoma cruzi circulating among dogs and triatomines in the
endemic countryside of the State of Rio Grande do Norte. Brazil
Acta Trop 200:105067. https:// doi. org/ 10. 1016/j. actat ropica. 2019.
105067
Ashford RW (1997) What it takes to be a reservoir host. Belg J Zool
127:85–90
Barbosa-Silva AN, Câmara ACJ, Martins K, Nunes DF, Oliveira PIC,
Azevedo PRM, Chiari E, Galvão LMC (2016) Characteristics of
triatomine infestation and natural Trypanosoma cruzi infection in
the State of Rio Grande do Norte, Brazil. Rev Soc Bras Med Trop
49:57–67. https:// doi. org/ 10. 1590/ 0037- 8682- 0300- 2015
Barbosa-Silva AN, Souza RCM, Diotaiuti L, Aguiar LMA, Câmara
ACJ, Galvão LMC, Chiari E (2019) Synanthropic triatomines
(Hemiptera: Reduviidae): infestation, colonization, and natural
infection by trypanosomatids in the State of Rio Grande do
Norte. Brazil Rev Soc Bras Med Trop 52:e20190061. https://
doi. org/ 10. 1590/ 0037- 8682- 0061- 2019
Barbosa-Silva AN (2017) Avaliação do risco de transmissão do
Trypanosoma cruzi no estado do Rio Grande do Norte: estudos
epidemiológicos e taxonômicos de triatomíneos (Hemiptera:
Reduviidae: Triatominae). Dissertation, Federal University of
Minas Gerais
Batista JS, Moura GHF, Lopes FC, Paiva KAR, Araújo Junior HN,
Góis RCS, Costa KMFM, Coelho WAC, Freitas CIA (2018) Risk
factors for trypanosomiasis by Trypanosoma vivax in cattle raised
Parasitology Research
1 3
in Rio Grande do Norte state. Arq Inst Biol 85:e0232016. https://
doi. org/ 10. 1590/ 1808- 16570 00232 016
Bezerra CM, Cavalcanti LPG, Souza RCM, Barbosa SE, Xavier SCC,
Jansen AM, Ramalho RD, Diotaiuti L (2014) Domestic, perido-
mestic and wild hosts in the transmission of Trypanosoma cruzi
in the Caatinga area colonised by Triatoma brasiliensis. Mem
Inst Oswaldo Cruz 109:887–898. https:// doi. org/ 10. 1590/ 0074-
02761 40048
Bezerra CM, Barbosa SE, Souza RCM, Barezani CP, Gürtler RE,
Ramos Júnior AN, Diotaiuti L (2018) Triatoma brasiliensis Neiva,
1911: food sources and diversity of Trypanosoma cruzi in wild
and artificial environments of the semiarid region of Ceará, north-
eastern Brazil. Parasit Vectors 11:642. https:// doi. org/ 10. 1186/
s13071- 018- 3235-4
Brito CRN, Sampaio GHF, Câmara ACJ, Nunes DF, Azevedo PRM,
Chiari E, Galvão LMC (2012) Seroepidemiology of Trypano-
soma cruzi infection in the semiarid rural zone of the State of Rio
Grande do Norte, Brazil. Rev Soc Bras Med Trop 45:346–352.
https:// doi. org/ 10. 1590/ S0037- 86822 01200 03000 13
Bryan LK, Hamer SA, Shaw S, Curtis-Robles R, Auckland LD, Hodo
CL, Chaffin K, Rech RR (2016) Chagas disease in a Texan horse
with neurologic deficits. Vet Parasitol 216:13–17. https:// doi. org/
10. 1016/j. vetpar. 2015. 11. 016
Câmara ACJ, Varela-Freire AA, Valadares HMS etal (2010) Genetic
analyses of Trypanosoma cruzi isolates from naturally infected tri-
atomines and humans in northeastern Brazil. Acta Trop 115:205–
211. https:// doi. org/ 10. 1016/j. actat ropica. 2010. 03. 003
Câmara ACJ, Lages-Silva E, Sampaio GHF, D’ Avila DA, Chiari E,
Galvão LMG (2013) Homogeneity of Trypanosoma cruzi I, II, and
III populations and the overlap of wild and domestic transmission
cycles by Triatoma brasiliensis in northeastern Brazil. Parasitol
Res 112:1543–1550. https:// doi. org/ 10. 1007/ s00436- 013- 3301-y
Carbajal-de-la-Fuente AL, Provecho YM, Fernández MP, Cardinal MV,
Lencina P, Spillmann C, Gürtler RE (2017) The eco-epidemiology
of Triatoma infestans in the temperate Monte Desert ecoregion of
mid-western Argentina. Mem Inst Oswaldo Cruz 112:698–708.
https:// doi. org/ 10. 1590/ 0074- 02760 160519
Cecere MC, Gurtler RE, Chuit R, Cohen JE (1997) Effects of chickens
on the prevalence of infestation and population density of Tria-
toma infestans in rural houses of north-west Argentina. Med Vet
Entomol 11:383–388. https:// doi. org/ 10. 1111/j. 1365- 2915. 1997.
tb004 26.x
Chagas C (1909) Nova tripanozomiase humana: estudos sobre a mor-
folojia e o ciclo evolutivo do Schizotrypanum cruzi n. gen., n. sp.,
ajente etiolojico de nova entidade morbida do homem. Mem Inst
Oswaldo Cruz 1:159–218
Chartier DI, Crocco LB (2007) Survey of Chagas disease vectors over
peridomiciles of the Ayacucho department rural area, province of
San Luis, Argentina. Rev Soc Entomol Argent 66:181–185
Cohen JE, Gürtler RE (2001) Modeling household transmission of
American trypanosomiasis. Science 293:694–698. https:// doi. org/
10. 1126/ scien ce. 10606 38
Colpo CB, Monteiro SG, Stainki DR, Colpo ETB, Henriques GB
(2005) Natural infection by Trypanosoma evansi in dogs. Cienc
Rural 35:717–719. https:// doi. org/ 10. 1590/ S0103- 84782 00500
03000 38
Costa SCL, Freitas JS, Silva AN, Lacerda LC, Cruz RDS, Carvalho FS,
Pereira MJS, Munhoz AD (2019) Frequency and factors associ-
ated with Theileria equi, Babesia caballi and Trypanosoma evansi
in equids from Bahia (Northeast Brazil). Rev Bras Parasitol Vet
28:47–58. https:// doi. org/ 10. 1590/ S1984- 29612 01800 90
Dias JCP, Machado EMM, Fernandes AL, Vinhaes MC (2000) Gen-
eral situation and perspectives of Chagas disease in Northeastern
Region, Brazil. Cad Saúde Pública 16:13–34. https:// doi. org/ 10.
1590/ S0102- 311X2 00000 08000 03
Drozino RN, Otomura FH, Gazarini J, Gomes ML, Toledo MJO (2019)
Trypanosoma Found in Synanthropic Mammals from Urban
Forests of Paraná, Southern Brazil. Vector-Borne Zoonotic Dis
19:828–834. https:// doi. org/ 10. 1089/ vbz. 2018. 2433
Fernandes AJ, Diotaiuti L, DiasJCP RomanhaAJ, Chiari E (1989)
Natural infection of the anal glands of the opossum (Didelphis
albiventris) by Trypanosoma cruzi in the municipality of Bam-
buí–MG, Brazil. Mem Inst Oswaldo Cruz 84:87–93. https:// doi.
org/ 10. 1590/ s0074- 02761 98900 01000 16
Filgueiras A, Barros JHS, Xavier SCC, Souza SF, Medeiros LS,
Ribeiro VMF, Jansen AM, Roque ALR (2019) Natural Trypano-
soma (Trypanozoon) evansi (Steel, 1885) infection among mam-
mals from Brazilian Amazon. Acta Trop 190:92–98. https:// doi.
org/ 10. 1016/j. actat ropica. 2018. 11. 011
Fuentes-Castillo A, Marzochi MCA, Modena CM, Silva VL, Bandeira
D, Judah A (1988) Epidemiological survey of human and caprine
infection by Trypanosoma cruzi in a rural area of the state of
Paraíba. Mem Inst Oswaldo Cruz 83:196
Garcia HA, Rodrigues CMF, Rodrigues AC, Pereira DL, Pereira CL,
Camargo EP, Hamilton PB, Teixeira MMG (2018) Remarkable
richness of trypanosomes in tsetse flies (Glossina morsitans mor-
sitans and Glossina pallidipes) from the Gorongosa National
Park and Niassa National Reserve of Mozambique revealed by
fluorescent fragment length barcoding (FFLB). Inf Gen Evol
63:370–379. https:// doi. org/ 10. 1016/j. meegid. 2017. 07. 005
Gomes ML, Macedo AM, Vago AR, Pena SDJ, Galvão LMC, Chiari
E (1998) Trypanosoma cruzi: optimization of polymerase chain
reaction for detection in human blood. Exp Parasitol 88:28–33.
https:// doi. org/ 10. 1006/ expr. 1998. 4191
Guerra RMSNC, Feitosa Júnior AB, Santos HP, Abreu-Silva AL, San-
tos ACG (2008) Biometry of Trypanosoma vivax found in a calf
in the state of Maranhão, Brazil. Cienc Rural 38:833–835. https://
doi. org/ 10. 1590/ S0103- 84782 00800 03000 41
Guindon S, Dufayard JF, Lefort V, Anisimova M, Hordijk W, Gascuel
O (2010) New algorithms and methods to estimate maximum-
likelihood phylogenies: assessing the performance of PhyML 3.0.
Syst Biol 59:307–321. https:// doi. org/ 10. 1093/ sysbio/ syq010
Gürtler RE, Cecere MC, Vázquez-Prokopec GM, Ceballos LA, Gure-
vitz JM, Fernández MP, Kitron U, Cohen JE (2014) Domestic
animal hosts strongly influence human-feeding rates of the Chagas
disease vector Triatoma infestans in Argentina. PLoS Negl Trop
Dis 8:e2894. https:// doi. org/ 10. 1371/ journ al. pntd. 00028 94
Herrera L, Morocoima A, Lozano-Arias D, García-Alzate R, Viettri M,
Lares M, Ferrer E (2022) Infections and coinfections by trypano-
somatid parasites in a rural community of Venezuela. Acta Para-
sitol 67:1015–1023. https:// doi. org/ 10. 1007/ s11686- 021- 00505-1
Honorato NRM, Barbosa-Silva AN, Negreiros CCA etal (2021) Tri-
atomine and Trypanosoma cruzi discrete typing units distribution
in a semi-arid area of northeastern Brazil. Acta Trop 220:105950.
https:// doi. org/ 10. 1016/j. actat ropica. 2021. 105950
IBGE – Instituto Brasileiro de Geografia e Estatistica (2021) Anuário
Estatístico do Brasil. http:// www. ibg e. gov. br/ cidad esat/ ufs/ downl
oad/ mapa_e_ munic ipios. php? ufrn. Accessed 10 March 2022
IDEMA – Instituto de Desenvolvimento Sustentável do Rio Grande do
Norte (2020) Anuário estatístico do Rio Grande do Norte 2020.
http:// www. idema. rn. gov. br using socioeconomico/anuario estatís-
tico2020. Accessed 10 March 2022
Ikenga JO, Richerson JV (1984) Trypanosoma cruzi (Chagas)(Protozoa:
Kinetoplastida: Trypanosomatidae) in invertebrate and vertebrate
hosts from Brewster County in Trans-Pecos Texas. J Econ Entomol
77:126–129. https:// doi. org/ 10. 1093/ jee/ 77.1. 126
Jansen AM, Xavier SCC, Roque ALR (2018) Trypanosoma cruzi
transmission in the wild and its most important reservoir hosts
in Brazil. Parasites Vectors 11:502. https:// doi. org/ 10. 1186/
s13071- 018- 3067-2
Parasitology Research
1 3
Jansen AM, Roque ALR (2010) Domestic and wild mammalian reser-
voir. In: Telleria J, Tibayrenc M (eds) American trypanosomiasis
Chagas disease-100 years of research, 2nd edn. Elsevier, London,
pp 249–276. https:// doi. org/ 10. 1016/ B978-0- 12- 384876- 5. 00011-3
Katoh K, Standley DM (2013) MAFFT multiple sequence alignment
software version 7: improvements in performance and usability.
Mol Biol Evol 30:772–780. https:// doi. org/ 10. 1093/ molbev/ mst010
Kumar S, Stecher G, Li M, Knyaz C, Tamura K (2018) MEGA X: molec-
ular evolutionary genetics analysis across computing platforms. Mol
Biol Evol 35:1547–1549. https:// doi. org/ 10. 1093/ molbev/ msy096
Lent H, Wygodzinsky PW (1979) Revision of the Triatominae (Hemip-
tera, Reduviidae), and their significance as vectors of Chagas’ dis-
ease. Bull Am Mus Nat Hist 163:127–520
Lilioso M, Reigada C, Pires-Silva D etal (2020) Dynamics of food
sources, ecotypic distribution and Trypanosoma cruzi infection in
Triatoma brasiliensis from the northeast of Brazil. PLoS Neg Trop
Dis 14:1–17. https:// doi. org/ 10. 1371/ journ al. pntd. 00087 35
Lima-Neiva V, Toma HK, Aguiar LMA, Lopes CM, Dias LP, Gonçalves
TCM, Costa J (2021) The connection between Trypanosoma cruzi
transmission cycles by Triatoma brasiliensis brasiliensis: A threat to
human health in an area susceptible to desertification in the Seridó,
Rio Grande do Norte. Brazil Plos Negl Trop Dis 15:e0009919.
https:// doi. org/ 10. 1371/ journ al. pntd. 00099 19
Martins K, Andrade CM, Barbosa-Silva AN, Nascimento GB, Chiari E,
Galvão LMC, Câmara ACJ (2015) Trypanosoma cruzi III causing
the indeterminate form of Chagas disease in a semi-arid region of
Brazil. Int J Inf Dis 39:68–75. https:// doi. org/ 10. 1016/j. ijid. 2015.
08. 012
Minh BQ, Nguyen MAT, Von Haeseler A (2013) Ultrafast approximation
for phylogenetic bootstrap. Mol Biol Evol 30:1188–1195. https:// doi.
org/ 10. 1093/ molbev/ mst024
Muñoz-San Martín C, Arbocco FCV, Saavedra M etal (2020) High rates
of Trypanosoma cruzi infection in goats from Mendoza province,
Argentina: parasite loads in blood and seasonal variation. Acta Trop
208:105493. https:// doi. org/ 10. 1016/j. actat ropica. 2020. 105493
Nguyen LT, Schmidt HA, Von Haeseler A, Minh BQ (2015) IQ-TREE:
a fast and effective stochastic algorithm for estimating maximum-
likelihood phylogenies. Mol Biol Evol 32:268–274. https:// doi. org/
10. 1093/ molbev/ msu300
Noyes HA, Stevens JR, Teixeira M, Phelan J, Holz P (1999) A nested
PCR for the ssrRNA gene detects Trypanosoma binneyi in theplaty-
pus and Trypanosoma sp. inwombatsandkangaroos in Australia. Int
J Parasitol 29:331–339. https:// doi. org/ 10. 1016/ s0020- 7519(98)
00167-2
Oliveira-Lima JW, Filho OFF, Vieira JBF, Gadelha FV, Oliveira-Filho
AM (2000) Peridomiciliary changes and implications for Triatoma
brasiliensis control. Cad Saúde Públia 16:75–81. https:// doi. org/ 10.
1590/ S0102- 311X2 00000 08000 08
PAHO (2022) Chagas disease. https:// www. paho. org/ en/ topics/ chagas-
disea se. Acessed 27 January 2022
Rodrigues A, Fighera RA, Souza TM, Schild AL, Soares MP, Milano J,
Barros CSL (2005) Surtos de tripanossomíase por Trypanosoma
evansi em equinos no Rio Grande do Sul: aspectos epidemiológicos,
clínicos, hematológicos e patológicos. Pesq Vet Bras 25:239–249.
https:// doi. org/ 10. 1590/ S0100- 736X2 00500 04000 10
Rodrigues MS, Lima L, Xavier SCC, Herrera HM, Rocha FL, Roque
ALR, Teixeira MMG, Jansen AM (2019) Uncovering Trypanosoma
spp. diversity of wild mammals by the use of DNA from blood
clots. Int J Parasitol Wildl 8:171–181. https:// doi. org/ 10. 1016/j. ijp-
paw. 2019. 02. 004
Ronquist F, Teslenko M, Van der Mark P etal (2012) MrBayes 3.2: effi-
cient Bayesian phylogenetic inference and model choice across a
large model space. Syst Biol 61:539–542. https:// doi. org/ 10. 1093/
sysbio/ sys029
Santos FR, Pena SDJ, Epplen JT (1993) Genetic and population study of
a Y-linked tetranucleotide repeat DNA polymorphism with a simple
non-isotopic technique. Hum Genet 90:655–656. https:// doi. org/ 10.
1007/ BF002 02486
Schenone H, Contreras MC, Borgoño JM, Maturana R, Salínas P, San-
doval L, Rojas A, Tello P, Villarroel F (1991) Overview of the epi-
demiology of Chagas’ disease in Chile. Bol Chi Parasitol 46:19–30
Smith A, Clark P, Averis S, Lymbery AJ, Wayne AF, Morris KD, Thomp-
son RCA (2008) Trypanosomes in a declining species of threatened
Australian marsupial, the brush-tailed bettong Bettongia penicillata
(Marsupialia: Potoroidae). Parasitology 135:1329–1335. https:// doi.
org/ 10. 1017/ S0031 18200 80048 24
Valença-Barbosa C, Fernandes FA, Santos HLC, Sarquis O, Harry M,
Almeida CE, Lima MM (2015) Molecular identification of food
sources in triatomines in the brazilian Northeast: roles of goats
and rodents in chagas disease epidemiology. Am J Trop Med Hyg
93:994–997. https:// doi. org/ 10. 4269/ ajtmh. 15- 0156
Vallejo GA, Guhl F, Chiari E, Macedo AM (1999) Species specific detec-
tion of Trypanosoma cruzi and Trypanosoma rangeli in vector and
mammalian hosts by polymerase chain reaction amplification of
kinetoplast minicircle DNA. Acta Trop 72:203–212. https:// doi.
org/ 10. 1016/ s0001- 706x(98) 00085-0
Vargas A, Malta JMAS, Costa VM, Cláudio LDG, Alves RV, Cordeiro
GS, Aguiar LMA, Percio J (2018) Investigation of an outbreak of
acute Chagas disease outside the Amazon Region, in Rio Grande
do Norte State, Brazil, 2016. Cad De Saúde Pública 34:e00006517.
https:// doi. org/ 10. 1590/ 0102- 311X0 00065 17
Wehrendt DP, Gómez-Bravo A, Ramirez JC etal (2019) Development
and evaluation of a duplex TaqMan qPCR assay for detection and
quantification of Trypanosoma cruzi infection in domestic and syl-
vatic reservoir hosts. Parasites Vectors 12:567. https:// doi. org/ 10.
1186/ s13071- 019- 3817-9
WHO (2022) Chagas disease (American trypanosomiasis). https:// www.
who. int/ health- topics/ chagas- disea se. Acessed 27 January 2022
Woo PTK (1970) The haematocrit centrifugation technique for the diag-
nosis of African trypanosomiasis. Acta Trop 27:384–386
Yefi-Quinteros E, Muñoz-San Martín C, Bacigalupo A, Correa JP, Cattan
PE (2018) Trypanosoma cruzi load in synanthropic rodents from
rural areas in Chile. Parasites Vectors 11:171. https:// doi. org/ 10.
1186/ s13071- 018- 2771-2
Zhang D, Gao F, Jakovlić I, Zou H, Zhang J, Li WX, Wang GT (2020)
PhyloSuite: An integrated and scalable desktop platform for stream-
lined molecular sequence data management and evolutionary phy-
logenetics studies. Mol Ecol Resour 20:348–355. https:// doi. org/ 10.
1111/ 1755- 0998. 13096
Publisher's note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
Article
Full-text available
An outbreak of Chagas disease, possibly involving its vector Triatoma brasiliensis brasilien-sis, was identified in the state of Rio Grande do Norte (RN). Given the historical significance of this vector in public health, the study aimed to evaluate its role in the transmission dynamics of the protozoan Trypanosoma cruzi in an area undergoing desertification in the Seridó region, RN, Brazil. We captured triatomines in sylvatic and anthropic ecotopes. Natural vector infection was determined using parasitological and molecular methods and we identified discrete typing units (DTUs) of T. cruzi by analyzing the COII gene of mtDNA, 24Sα rDNA, and mini-exon gene. Their blood meals sources were identified by amplification and sequencing of the mtDNA cytochrome b gene. A total of 952 T. b. brasiliensis were captured in peridomestic (69.9%) and sylvatic ecotopes (30.4%). A wide range of natural infection rates were observed in peridomestic (36.0%-71.1%) and sylvatic populations (28.6%-100.0%). We observed the circulation of TcI and TcII DTUs with a predominance of Tcl in sylvatic and peridomestic environments. Kerodon rupestris, rocky cavy (13/39), Homo sapi-ens, human (8/39), and Bos taurus, ox (6/39) were the most frequently detected blood meals sources. Thus, Triatoma b. brasiliensis is invading and colonizing the human dwellings. Furthermore, high levels of natural infection, coupled with the detection of TcI and TcII DTUs, and also the detection of K. rupestris and H. sapiens as blood meals sources of infected T. b. brasiliensis indicate a risk of T. cruzi transmission to human populations in areas undergoing desertification.
Article
Full-text available
Innovative approaches used to combat Chagas disease transmission tend to combine a set of comprehensive efforts to understand the ecology of local vectors. In this work we identified molecularly the blood meal of 181 Triatoma brasiliensis, distributed in 18 populations (8 sylvatic and 10 peridomestic), which were collected across a range of 240 km (East-West) and 95 km (North-South) in the semi-arid region of northeastern, Brazil. We used the vertebrate mitochondrial gene (cytochrome B) sequencing applied to DNA isolated from bug mid-gut to identify the insect blood meal sources via the BLAST procedure. The peridomestic populations were classified according to two main hypotheses of site-occupancy for T. brasi-liensis: the first says that the infestation is mainly driven by structures that resemble its natural habitat (stony-like ecotopes) and the second assumes that it is associated with key-hosts (rodents and goats). Rodents of the Caviidae family (Galea spixii and Kerodon rupestris) were identified as the key-host of T. brasiliensis, but also the potential Trypanosoma cruzi reservoir-able to connect the sylvatic and domestic T. cruzi cycle. Cats also deserve to be studied better, as potential T. cruzi reservoirs. By modeling the food sources + site-occupancy + T. cruzi natural infection, we identified man-made ecotopes suitable for forming dense triatomine infestations with high rates of T. cruzi natural infection, which may be taken into account for vector control measures.
Article
Full-text available
Background: A question of epidemiological relevance in Chagas disease studies is to understand Trypanosoma cruzi transmission cycles and trace the origins of (re)emerging cases in areas under vector or disease surveillance. Conventional parasitological methods lack sensitivity whereas molecular approaches can fill in this gap, provided that an adequate sample can be collected and processed and a nucleic acid amplification method can be developed and standardized. We developed a duplex qPCR assay for accurate detection and quantification of T. cruzi satellite DNA (satDNA) sequence in samples from domestic and sylvatic mammalian reservoirs. The method incorporates amplification of the gene encoding for the interphotoreceptor retinoid-binding protein (IRBP), highly conserved among mammalian species, as endogenous internal amplification control (eIAC), allowing distinction of false negative PCR findings due to inadequate sample conditions, DNA degradation and/or PCR interfering substances. Results: The novel TaqMan probe and corresponding primers employed in this study improved the analytical sensitivity of the assay to 0.01 par.eq/ml, greater than that attained by previous assays for Tc I and Tc IV strains. The assay was tested in 152 specimens, 35 from 15 different wild reservoir species and 117 from 7 domestic reservoir species, captured in endemic regions of Argentina, Colombia and Mexico and thus potentially infected with different parasite discrete typing units. The eIACs amplified in all samples from domestic reservoirs from Argentina and Mexico, such as Canis familiaris, Felis catus, Sus scrofa, Ovis aries, Equus caballus, Bos taurus and Capra hircus with quantification cycles (Cq's) between 23 and 25. Additionally, the eIACs amplified from samples obtained from wild mammals, such as small rodents Akodon toba, Galea leucoblephara, Rattus rattus, the opossums Didelphis virginiana, D. marsupialis and Marmosa murina, the bats Tadarida brasiliensis, Promops nasutus and Desmodus rotundus, as well as in Conepatus chinga, Lagostomus maximus, Leopardus geoffroyi, Lepus europaeus, Mazama gouazoubira and Lycalopex gymnocercus, rendering Cq's between 24 and 33. Conclusions: This duplex qPCR assay provides an accurate laboratory tool for screening and quantification of T. cruzi infection in a vast repertoire of domestic and wild mammalian reservoir species, contributing to improve molecular epidemiology studies of T. cruzi transmission cycles.
Article
Full-text available
Multigene and genomic data sets have become commonplace in the field of phylogenetics, but many existing tools are not designed for such data sets, which often makes the analysis time‐consuming and tedious. Here, we present PhyloSuite, a (crossplatform, open‐source, stand‐alone Python graphical user interface) user‐friendly workflow desktop platform dedicated to streamlining molecular sequence data management and evolutionary phylogenetics studies. It uses a plugin‐based system that integrates several phylogenetic and bioinformatic tools, thereby streamlining the entire procedure, from data acquisition to phylogenetic tree annotation (in combination with iTOL). It has the following features: (a) point‐and‐click and drag‐and‐drop graphical user interface; (b) a workplace to manage and organize molecular sequence data and results of analyses; (c) GenBank entry extraction and comparative statistics; and (d) a phylogenetic workflow with batch processing capability, comprising sequence alignment (mafft and macse), alignment optimization (trimAl, HmmCleaner and Gblocks), data set concatenation, best partitioning scheme and best evolutionary model selection (PartitionFinder and modelfinder), and phylogenetic inference (MrBayes and iq‐tree). PhyloSuite is designed for both beginners and experienced researchers, allowing the former to quick‐start their way into phylogenetic analysis, and the latter to conduct, store and manage their work in a streamlined way, and spend more time investigating scientific questions instead of wasting it on transferring files from one software program to another.
Article
Full-text available
Introduction: The ecoepidemiological situation in the State of Rio Grande do Norte, Brazil is characterized by frequent invasion and colonization of domiciliary units (DUs) by several triatomine species, with high rates of natural infection by Trypanosoma cruzi. METHODS: We evaluated the possibility of vector transmission of T. cruzi based on records of the occurrence of domiciled triatomines collected by the Secretariat of State for Public Health from 2005 to 2015. During this period, 67.7% (113/167) of municipalities conducted at least one active search and 110 recorded the presence of insects in DUs. These activities were more frequent in municipalities considered to have a high and medium-level risk of T. cruzi transmission. Results: Of 51,569 captured triatomines, the most common species were Triatoma brasiliensis (47.2%) and T. pseudomaculata (40.2%). Colonies of T. brasiliensis, T. pseudomaculata, T. petrocchiae, Panstrongylus lutzi, and Rhodnius nasutus were also recorded in the intradomicile and peridomicile. Natural infection by trypanosomatids was detected in 1,153 specimens; the highest rate was found in R. nasutus (3.5%), followed by T. brasiliensis (2.5%) and T. pseudomaculata (2.4%). There have been high levels of colonization over the years; however, not all infested DUs have been sprayed. CONCLUSIONS: This is the first report of intradomicile and peridomicile colonization by P. lutzi. These results demonstrate the risk of new cases of infection by T. cruzi and reinforce the need for continuous entomological surveillance in the State of Rio Grande do Norte.
Article
Full-text available
Trypanosoma cruzi is a parasitic protozoan that infects a diversity of hosts constituting the cycle of enzootic transmission in wild environments and causing disease in humans (Chagas disease) and domestic animals. Wild mammals constitute natural reservoirs of this parasite, which is transmitted by hematophagous kissing bugs of the family Reduviidae. T. cruzi is genetically subdivided into six discrete typing units (DTUs), T. cruzi (Tc)I to TcVI. In Brazil, especially in the state of Paraná, TcI and TcII are widely distributed. However, TcII is less frequently found in wild reservoirs and triatomine, and more frequently found in patients. The goal of this study was to investigate the natural occurrence of T. cruzi in wild synanthropic mammals captured in urban forest fragments of the Atlantic Forest of Paraná, southern Brazil. In this way, 12 opossums and 35 bats belonging to five species were captured in urban forest parks of the city of Maringá, Paraná, an area considered endemic for Chagas disease. PCR-kinetoplast DNA molecular diagnostic reveals Trypanosoma sp. infection in 12 (100%) Didelphis albiventris and 10 (40%) Artibeus lituratus. In addition to demonstrating the presence of Trypanosoma in the two groups of mammals studied, we obtained an isolate of the parasite genotyped as TcII by amplification of the cytochrome oxidase II gene by PCR, followed by restriction fragment length polymorphism with AluI, and confirmed by PCR of rDNA 24Sα. This is the first record of the encounter in wild mammals of Trypanosoma DNA (in A. lituratus) and T. cruzi DTU TcII (in D. albiventris) in the state of Paraná.
Article
Introduction: Trypanosoma cruzi, Trypanosoma rangeli and Leishmania spp. are parasites that coexist in several endemic areas. The identification of these parasites in hosts is important for the control programs. Methods: 216 samples from human blood (101), blood of other mammals (45) and triatomine intestinal content and hemolymph (70), from an endemic area of Venezuela, were analysed. The samples were evaluated by; serology (only humans) and PCR for T. cruzi in human, other mammals and triatomines, PCR for T. rangeli in mammals-including human and triatomines and PCR for Leishmania in mammals-including human. Results: The 9.9% of the human samples were positive for T. cruzi by serology, 11.9% by PCR, 4% for T. rangeli PCR and none for Leishmania spp. PCR. 60% of the samples of other mammals showed DNA amplification for T. cruzi, 42.2% for T. rangeli and 4.4% for Leishmania spp. 61.4% of the triatomine samples showed DNA amplification for T. cruzi and 10% for T. rangeli. Conclusions: High T. cruzi infection was detected in mammals and triatomines compared with T. rangeli. Low leishmanial infection was detected in other mammals. It is the first time that T. cruzi/T. rangeli coinfection, in humans, Canis familiaris (dog), and Bos Taurus (cow), were reported world-wide, and that this coinfection was described in Tamandua tetradactyla (anteater) from Venezuela. The coinfection T. cruzi/T. rangeli in mammals-including humans and triatomines, and coinfection T. cruzi/Leishmania spp. in non-human mammals, show the risk for trypanosomic zoonoses in this endemic area.
Article
The occurrence of triatomine species, their bloodmeal sources, and the discrete typing units (DTUs) of Trypanosoma cruzi isolated from them were determined in different municipalities of the state of Rio Grande do Norte, Brazil. Triatomine captures were carried out in the rural areas of 23 municipalities. The genotyping of T. cruzi isolates was performed using the mitochondrial cytochrome c oxidase subunit 2 (coii) gene, the D7 region of the 24Sα rDNA, and the spliced leader intergenic region (SL-IR). Five triatomine species were captured, and the most frequent was Triatoma brasiliensis (84.3%; 916/1086), which was found in 16 of the 23 municipalities surveyed, and infested all types of environment investigated. The TcI DTU was found in all mesoregions surveyed in 51.5% (17/33) of the culture-positive samples. In contrast, TcII (9.1%; 3/33) was detected in the Central mesoregion, while TcIII (27.3%; 9/33) was found in all mesoregions. The geographic distribution and spatial overlap of different DTUs was inferred using the superposition of the radius of occurrence of isolates and using ecological niche distribution modelling. Triatoma brasiliensis was found infected in all mesoregions and with all three T. cruzi DTUs, including mixed infections. With regard to bloodmeal sources, the DNA of rodents was found in triatomines infected with either TcI or TcIII, while that of domestic animals and humans was associated with both single and mixed infections. Our findings demonstrate that different DTUs of T. cruzi are widely dispersed among triatomines in our study area. The association of T. brasiliensis with several different mammalian hosts, as well as overlapping areas with different DTUs, suggests that this triatomine species may have an important role as a vector in both anthropic and sylvatic environments.
Article
Mendoza province, in central west Argentina, is considered among the high-risk provinces for vector transmission of Trypanosoma cruzi, the causative agent of Chagas disease. Extensive goat farming is common in large parts of rural Mendoza, and goats may act as a reservoir for this parasite. The objective of this study was to determine infection rates, parasite loads, and seasonal variation of these parameters in T. cruzi infection in goats from rural areas of three departments of Mendoza. A total of 349 peripheral blood samples with EDTA / guanidine were analyzed from goats on 11 farms (three in Lavalle, three in San Carlos, and five in Malargüe department) in spring of 2014, 2015, and 2016; and in fall of 2015 and 2016 (only Malargüe). DNA was extracted using a Phenol: Chloroform: Isoamyl protocol. The detection and quantification of T. cruzi was performed through qPCR amplification using satellite oligonucleotides. Of the 349 blood samples, 267 (77%) were positive, with parasite loads ranging between <0.10 and 10.90 par-eq/mL (median 0.10). In spring, frequencies of infection in the three departments ranged between 86% and 95%, but differences were not significant. Median parasite loads were higher in Lavalle than in the other departments, while those in goats from San Carlos were consistently low. The frequency of infection and parasite loads in Malargüe were significantly higher in spring than in fall. This seasonal variation may have been related to a reduced nutritional status and impaired immune response of goats in spring. In conclusion, the high proportion of positive goats confirms the persistence of T. cruzi in rural Mendoza.
Article
Vector transmission of Trypanosoma cruzi occurs in several areas of Brazil, including the northeastern region, and domestic animals can serve as reservoirs of the parasite. The aim of this study was to monitor dogs as domestic reservoirs for infection by T. cruzi, and the main triatomine species involved in parasite transmission in rural areas of municipalities in the State of Rio Grande do Norte, in northeastern, Brazil. Blood samples from dogs (n = 40) and manual triatomine capture were performed in domiciliary and peridomiciliary environments in rural areas of the towns of Acari, Caraúbas and Marcelino Vieira, between 2013 and 2016. Subsequently, infection of dogs was determined by Polymerase Chain Reaction (PCR), Enzyme-Linked Immunosorbent Assay (ELISA) for the detection of IgM and IgG isotypes and Indirect Immunofluorescence (IIF) reactions for detection of IgG. Triatomine infection was determined by PCR. Forty (16/40) percent of the dogs were seropositive for T. cruzi; 20.0% (8/40) of such reactivity indicated the acute phase, and 20.0% (8/40), the chronic phase. PCR was positive in 42.5% (17/40) of the dogs' blood samples. Specimens of Triatoma brasiliensis, Triatoma pseudomaculata, Rhodnius nasutus and Panstrongylus lutzi were found to be infected; however only T. brasiliensis nymphs and adults were infected in both environments. Triatomines evaluation showed 82.5% (94/114) of PCR positivity. Taken together, our results confirm that dogs are domestic reservoirs of T. cruzi in northeastern Brazil and T. brasiliensis is the main triatomine species correlated with parasite transmission in domiciliary environments. There is a continuing need to control peridomiciliary populations of triatomines and to implement continuous surveillance strategies for reservoirs with the help from the community.