You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/352222793

Molecular ecology of Triatoma dimidiata in southern Belize reveals risk for


human infection and the local differentiation of Trypanosoma cruzi parasites

Article  in  International journal of infectious diseases: IJID: official publication of the International Society for Infectious Diseases · June 2021
DOI: 10.1016/j.ijid.2021.05.083

CITATIONS READS

0 2

4 authors, including:

Jaime López Domínguez


Universidad Veracruzana
7 PUBLICATIONS   0 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Insect Immune response View project

All content following this page was uploaded by Jaime López Domínguez on 18 June 2021.

The user has requested enhancement of the downloaded file.


International Journal of Infectious Diseases 108 (2021) 320–329

Contents lists available at ScienceDirect

International Journal of Infectious Diseases


journal homepage: www.elsevier.com/locate/ijid

Molecular ecology of Triatoma dimidiata in southern Belize reveals


risk for human infection and the local differentiation of
Trypanosoma cruzi parasites
Roy Polonioa , Jaime López-Domínguezb,c,d , Claudia Herrerab , Eric Dumonteilb,*
a
University of Belize, Punta Gorda, Toledo, Belize
b
Department of Tropical Medicine, School of Public Health and Tropical Medicine, Vector-Borne and Infectious Disease Research Center, Tulane University,
New Orleans, LA, USA
c
LADISER Inmunología y Biología Molecular, Facultad de Ciencias Químicas, Universidad Veracruzana, Orizaba, Veracruz, Mexico
d
Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, Veracruz, Mexico

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

Article history: Objective: In Belize, the main vector for Trypanosoma cruzi, the agent of Chagas disease, is Triatoma
Received 21 April 2021 dimidiata, but transmission cycles and the risk for human infection are unclear. Therefore, the aim of this
Received in revised form 21 May 2021 study was to identify T. dimidiata blood feeding sources and its parasite and microbial diversity, in order
Accepted 31 May 2021
to reconstruct T. cruzi parasite transmission ecology in southern Belize.
Methods: A metabarcoding approach based on deep sequencing of markers was used for bug taxonomy,
Keywords: blood meal sources, T. cruzi genotypes, and microbiota composition. Bugs were collected in 13 villages of
Hematophagous
Toledo district.
Blood meal
Transmission network
Results: Bugs fed on at least 13 species, from domestic hosts such as humans, dogs, cows, and pigs, to
Chagas disease synanthropic species such as mice, rats, and opossums, and sylvatic species such as deer, peccary, and
Microbiome kinkajou, in agreement with an opportunistic feeding behavior. Nonetheless, most feeding focused on a
Parasite diversity few species, including humans. Infection with T. cruzi was detected in 24 of 39 bugs (62%), and the
analysis of 242 T. cruzi mini-exon sequences (average 10  5 haplotypes per bug) indicated the presence
of TcI and TcIV parasite discrete typing units (DTUs). However, for both DTUs, sequences from Belize
mostly clustered apart from sequences from North and South America, suggesting the local
differentiation of parasites. T. dimidiata also harbored a diverse bacterial microbiota, with ontogenic
changes suggesting microbiota maturation during nymphal development.
Conclusions: Together, these results indicate a significant risk for T. cruzi infection in humans. They also
highlight the need to better characterize the diversity of T. cruzi strains in the region and its impact on
disease epidemiology.
© 2021 The Author(s). Published by Elsevier Ltd on behalf of International Society for Infectious Diseases.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-
nd/4.0/).

Introduction situation is unclear. A seroprevalence of 2.7% was reported in Cayo


district in 1967, with some of the seropositive cases presenting
Chagas disease is a neglected tropical disease in the Americas cardiac arrhythmias (Coura and Petana, 1967). In 1997, a study of
caused by the vector-borne protozoan parasite Trypanosoma cruzi. several populations reported a variable seroprevalence ranging
There are an estimated 6–8 million infected persons in the from 0 to 6.1% (Jaramillo et al., 1997). Currently, the Ministry of
continent, resulting in a burden of 29 000 000 disability-adjusted Health of Belize considers that there are no confirmed cases of
life years (DALYs) and a health care cost of US$ 24.73 billion (Hotez Chagas disease, and maintains a continuous screening program for
et al., 2008, 2013; Lee et al., 2013). In Belize, the epidemiological blood donors (Belize Ministry of Health, 2014).
The main vector in Belize is Triatoma dimidiata, which was first
described in the country in 1932, and found widespread in sylvatic
areas, with limited domiciliation (Petana, 1972). Seasonal infesta-
* Corresponding author at: Department of Tropical Medicine, School of Public
tion of houses by T. dimidiata has been detected in multiple villages
Health and Tropical Medicine, Vector-Borne and Infectious Disease Research Center,
Tulane University, 1440 Canal St., New Orleans, LA 70112, USA. in Cayo and Toledo districts, suggesting some risk for human
E-mail address: edumonte@tulane.edu (E. Dumonteil). transmission (Polonio et al., 2009). Such a seasonal infestation

https://doi.org/10.1016/j.ijid.2021.05.083
1201-9712/© 2021 The Author(s). Published by Elsevier Ltd on behalf of International Society for Infectious Diseases. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

pattern is very similar to that observed in southern Mexico and microbial diversity, in order to reconstruct T. cruzi parasite
parts of Guatemala (Dumonteil et al., 2002, 2013; Waleckx et al., transmission ecology. A metabarcoding approach was used, based
2015b). It is associated with a human seroprevalence of at least 1– on deep sequencing of selected markers from bugs; this has proven
5% in Yucatan, Mexico (Gamboa-León et al., 2014; Buekens et al., powerful in reconstructing the ecological interactions within these
2018). In addition, T. dimidiata is rather a species complex, and complex parasite transmission cycles (Dumonteil et al., 2018,
several haplogroups have been described based on specific 2020a,b; Murillo-Solano et al., 2021).
markers, some of these potentially representing species or
subspecies (Dorn et al., 2009; Justi et al., 2018). However, the Methods
epidemiological relevance of these taxonomic distinctions remains
unclear, as some haplogroups and their hybrids are found in Study area and triatomine collection
sympatry in the Yucatan Peninsula of Mexico and follow a similar
seasonal house infestation pattern (Herrera-Aguilar et al., 2009). Toledo district is located in the southern part of Belize (Figure 1),
A high genetic diversity is also present in T. cruzi parasites, between latitude 15.8 and 16.7 North and longitude 88.3 and 89.2
which are currently classified into seven main discrete typing units West, and has a population of 35 800 inhabitants. Forty-two bugs
(DTUs) (Zingales et al., 2009, 2012). TcI was thought to predomi- derived from previous collections were analyzed (Polonio et al.,
nate in Mexico (Bosseno et al., 2002), but recent studies have 2009); these bugs were obtained from 13 rural villages across Toledo
detected several other DTUs (Ramos-Ligonio et al., 2012; Lopez- district (Figure 1), collected inside or around houses, and included
Cancino et al., 2015; Dumonteil et al., 2018; Villanueva-Lizama adults and nymphs (Supplementary material Table S1).
et al., 2019), and infections with TcII/TcV/TcVI may predominate in
humans (Herrera et al., 2019). In Belize, limited information is DNA extraction, PCR amplification of markers, and sequencing
available on T. cruzi diversity (Breniere et al., 2016). A study of
T. dimidiata vectors, including two from Toledo, detected TcI and The inside of the abdomen was washed twice with 0.2 ml of
TcIV parasites, and phylogenetic analysis grouped Central and sterile water using a 1-ml syringe, and DNA was extracted from the
North American TcI parasites in a single cluster (Dorn et al., 2017). wash solution using the Qiagen DNeasy Blood and Tissue Kit.
However, a significant geographic structuration of parasite strains Specific markers were amplified for the simultaneous identifica-
within DTUs is emerging (Llewellyn et al., 2009; Majeau et al., tion of T. dimidiata haplogroup, their vertebrate blood meals,
2021), pointing to the likely local differentiation of parasite strains, T. cruzi parasites, and gut microbiota, as described previously
which may be reflected in differences in transmission cycles or (Dumonteil et al., 2018, 2020a,b). DNA extraction, PCR reaction
pathogenesis. preparation, PCR amplification, and gel electrophoresis were
These observations raise the question of the nature of T. cruzi performed in separate laboratory rooms, to prevent cross-
transmission cycles in Belize and the potential for human contaminations, and all PCR reactions included positive and
infections. Therefore, the aim of this study was to identify T. negative controls. PCR products for all markers were pooled for
dimidiata blood feeding sources as well as its parasite and each bug and sequenced on a MiSeq (Illumina) platform. Raw

Figure 1. Map of the study area. (A) General map of southern Mexico and part of Central America. (B) Map of Toledo district. Villages for bug collections are indicated, and the
size of each yellow circle is proportional to their population. The insert shows the location of Toledo district in Belize.

321
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

sequence data are available from the NCBI SRA database discarded from the analysis. Phylogenetic analysis of ITS-2
under BioProject #PRJNA719290, BioSamples #SAMN18594627- sequences allowed the identification of the haplogroup present
SAMN18594665. (Figure 2). Most collected bugs belonged to haplogroup 2 (30/39),
followed by haplogroup 3 (5/39) and haplogroup 1A (1/39). Three
Sequence and data analysis bugs harbored ITS-2 sequences from haplogroups 2 and 3, and
were considered hybrids (Herrera-Aguilar et al., 2009).
Raw FASTQ sequences files were mapped to reference sequences
from each DNA marker. Sequence variants of the respective markers Blood feeding sources and parasite transmission networks
were identified using FreeBayes SNP/variant tool (Garrison and
Marth, 2012). Triatomine ITS-2 haplotypes were then aligned with Analysis of 12S RNA gene sequences allowed the identification
reference sequences from haplogroups 1A, 1B, 2, and 3 (Dorn et al., of blood feeding sources from 13 vertebrate species. These
2009) and a maximum likelihood phylogenetic tree was elaborated included domestic species such as dogs, cows, and pigs,
using PhyML as implemented in Geneious, with 100 bootstrap to synanthropic species such as mice, rats, and opossums, and
assess branch support. ITS-2 sequences were deposited in GenBank sylvatic species such as deer, peccary, and kinkajou, in agreement
under accession numbers MW856024–MW856063. with an opportunistic feeding behavior of T. dimidiata (Dumonteil
For blood meal sources, 12S sequences were analyzed by et al., 2018). Nonetheless, feeding was focused on a limited number
MegaBLAST against the ‘nr’ GenBank database. A sequence match of hosts, which accounted for the majority of blood meals. Analysis
with >97% identity was used for species/genus identification. of alpha diversity of feeding sources indicated that while male and
Sequences were further filtered to remove low abundance female bugs had a similar diversity of feeding sources, nymphs
sequences (<0.5% of reads) and normalized using the total sum tended to have a different feeding diversity, although this did not
scaling (TSS) method, to account for variability in sequencing reach statistical significance (F = 1.37, P = 0.27 and F = 1.38, P = 0.27
depth among samples. Alpha diversity (within bugs) of blood for Chao1 and Shannon indices, respectively) (Figure 3A, B).
feeding sources was assessed by calculating Chao1 and Shannon However, further comparison of feeding profiles indicated signifi-
indices, which were compared by analysis of variance (ANOVA). cant differences among nymphs and adult bugs, with nymphs
Beta diversity of feeding sources among T. dimidiata subpopula- feeding frequently on dogs rather than on humans, and adults
tions was analyzed by non-metric dimensional scaling (NMDS), feeding predominantly on humans rather than on dogs (PERMA-
based on Bray–Curtis index distance measures, and statistical NOVA; F = 4.31, P = 0.011) (Figure 3C, D). Finally, no significant
significance of differences in feeding hosts among groups was differences in blood meal sources were found among adult bugs
evaluated by permutational multivariate ANOVA (PERMANOVA). from the different ITS-2 haplogroups (PERMANOVA; F = 0.90;
Data on multiple blood meal sources in single bugs were used to P = 0.44), suggesting extensive sympatry and their involvement in a
construct feeding networks and parasite transmission pathways in common feeding network.
Cytoscape 3.8 (Dumonteil et al., 2018, 2020a,b). Multiple blood meal sources were detected in most bugs, with
Parasite sequences from the mini-exon gene were aligned with an average of 4.6  1.5 hosts per bug, confirming frequent host-
sequences from reference strains, and maximum-likelihood switching, and allowing the reconstruction of a feeding and
phylogenetic trees were constructed. Sequences were deposited parasite transmission network. The network was found to be
in the GenBank database under accession numbers MW861761– biased towards domestic species that are frequent blood meal
MW862002. Similarity among TcI sequences was further assessed
by principal component analysis using PAST 4.0 software (Hammer
et al., 2001). Statistical significance of differences among groups
was evaluated by PERMANOVA. Sequences from TcI and TcIV DTUs
were further analyzed with additional sequences from other
geographic regions. For this, only the two most frequent T. cruzi
haplotypes from each bug and DTU were included in the analyses.
Sequences were analyzed in Beast 2.6.2 (Bouckaert et al., 2019)
based on an HKY substitution model for a constant coalescent
population, and the molecular clock was set based on a
substitution frequency of 7.1  10 8 (Torres-Silva et al., 2018).
The model was run for 10 million iterations to obtain maximum
credibility trees.
For microbiota composition, bacterial 16S sequences were
analyzed using a Bayesian classifier from the Ribosomal Database
Project (Cole et al., 2014). Taxonomic identification of bacteria was
made at a threshold of >97% sequence identity of bacterial families.
Alpha diversity of the microbiota was assessed with Chao1 and
Shannon indices in Microbiome Analyst (Dhariwal et al., 2017),
which were compared by Student t-test or ANOVA. Beta diversity
was compared by NMDS, based on Bray–Curtis index distance
measures, and statistical significance of differences was evaluated
by PERMANOVA.

Results

T. dimidiata ITS-2 haplogroup diversity Figure 2. Phylogenetic analysis of Triatoma dimidiata ITS-2 sequences. Partial ITS-2
sequences were analyzed using maximum likelihood, and the bootstrap branch
support is indicated. Bugs from Belize (BEL) clustered with reference sequences
From the 42 analyzed bugs, good amplification of markers and from haplogroups 2, 1A, and 3. Three bugs (BEL160, BEL181, and BEL212) presented
quality sequences were obtained for 39 bugs and three were sequences from haplogroups 2 (-A) and 3 (-B), and were considered hybrids.

322
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

Figure 3. Blood feeding profiles of Triatoma dimidiata. Chao1 (A) and Shannon (B) alpha diversity indices were compared among males, females, and nymphs from Toledo
district, but differences did not reach statistical significance (F = 1.37, P = 0.27 and F = 1.38, P = 0.27, respectively). Feeding profiles differed between adults and nymphs (C), with
a significant difference in beta diversity according to developmental stage. (D) NMDS analysis of beta diversity (PERMANOVA; F = 4.31, P = 0.011).

sources in association with humans (Figure 4), as expected for bugs 10  5 haplotypes per bug). Phylogenetic analysis of these
collected inside/around houses. Chickens, although present in the sequences indicated that they clustered with TcI and TcIV DTUs,
feeding network, do not seem to be a major host and were the only while the other parasite DTUs were not detected in these bugs
avian species detected. Humans were a major node in the network (Figure 5A). TcI sequences were detected in 24 bugs, and two bugs
and their infection may have been derived from T. cruzi strains presented co-infections with TcIV. Phylogenetic comparison of TcI
present in multiple domestic animals such as dogs, cats, cows, and sequences indicated that some haplotypes were shared among
pigs, which are frequently associated in multiple blood meals, bugs, suggesting that identical/similar strains were infecting these
confirming a significant risk for human infections. In addition, bugs, while other haplotypes were found in single bugs (Figure 5B).
several sylvatic species were also involved in this network, Principal component analysis of TcI haplotypes indicated that there
emphasizing the connectivity between the sylvatic and domestic was no significant difference in mini-exon sequence diversity
habitats, in agreement with the seasonal infestation of houses by among T. dimidiata ITS-2 haplogroups (PERMANOVA; F = 1.34,
bugs from sylvatic areas (Dumonteil et al., 2007, 2009; Polonio P = 0.21) (Figure 5C), suggesting that similar parasites circulate
et al., 2009; Barbu et al., 2010). This connectivity may allow for among these bug subpopulations. On the other hand, significant
strains of T. cruzi infecting sylvatic animals to reach domestic hosts differences in TcI parasite haplotypes were found according to the
and humans. developmental stages of the bugs (PERMANOVA; F = 14.01,
P = 0.0001) (Figure 5D), likely in agreement with their different
T. cruzi parasite diversity feeding profiles. In addition, significant differences in TcI parasite
haplotypes were found among villages, pointing to geographic
The diversity of T. cruzi parasite strains in T. dimidiata was differences among TcI strains (PERMANOVA; F = 18.79, P = 0.011)
assessed by the mini-exon marker, which provides extensive (Figure 5E).
information on genetic diversity both among and within parasite TcI and TcIV sequences from Belize were then compared with
DTUs (Majeau et al., 2019, 2021). Infection with T. cruzi was sequences from other geographic regions in further phylogenetic
detected in 24 of 39 bugs (62%), corresponding to 19 of 30 (63%) for analyses. For TcIV sequences, these clearly differed from the South
ITS-2 haplogroup 2, three of five (60%) for haplogroup 3, one of American CanIII reference, but they also clustered separately from
three (33%) for hybrids (haplogroups 2/3), and one of one (100%) sequences from the USA, indicating the local differentiation of the
for haplogroup 1A. A total of 242 mini-exon sequences were TcIV strains from Belize (Figure 6A). For TcI strains, sequences from
obtained, ranging from 1 to 21 sequences per infected bug (average Belize clustered in a group of sequences that mostly included

323
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

Figure 4. Triatoma dimidiata blood feeding and Trypanosoma cruzi parasite transmission network. This network integrates feeding frequency on the distinct hosts and the co-
occurrence of hosts in multiple blood meals from individual bugs. The size of vertebrate hosts nodes is proportional to the frequency of blood meals on each host, and the
width of the edges connecting hosts is proportional to the frequency of their co-occurrence in single bug blood meals. Solid edges link mammalian hosts (green circles) that
are competent hosts for T. cruzi, while dotted edges link non-competent hosts (birds: blue hexagons).

sequences from Panama, although a few sequences from Colombia microbiota composition according to the developmental stage of
and the USA were also present, pointing to the local differentiation the bugs (PERMANOVA; F = 1.82, P = 0.04). In nymphs, Staph-
of TcI strains in Central America (Figure 6B). Most of the sequences ylococcaceae, Brevibacteriaceae, and Bacillaceae comprised the large
from the USA and Mexico were found in a distinct cluster, majority of bacteria present, while in adult males and females a
previously identified as belonging to the TcIa subgroup, while larger diversity of bacterial families was detected (Figure 7C, D).
sequences from Colombia belonging to the TcIb subgroup formed In adult bugs, microbiota composition among ITS-2 hap-
another cluster, as well as sequences from South America logroups tended to be different (Figure 8A), but this did not reach
corresponding to subgroups TcId and TcIe. These results indicate statistical significance (PERMANOVA; F = 0.55, P = 0.84), likely
that TcI and TcIV sequences from Belize are distinct from sequences because of limited sample sizes. Similarly, microbiota composition
from North and South America. seemed to vary according to T. cruzi infection status of ITS-2
haplogroups 2 and 3 bugs, without reaching statistical significance
T. dimidiata gut microbiota (PERMANOVA; F = 0.38, P = 0.94 and F = 0.52, P = 0.9 for
haplogroups 2 and 3, respectively) (Figure 8B, C). Nonetheless,
Analysis of bacterial 16S sequence indicated a diverse microbial Bacillaceae, Moraxellaceae, and Enterobacteriaceae were more
diversity in these bugs, with over 36 microbial families detected at abundant in uninfected bugs from haplogroup 2 compared to
a frequency of at least 0.5%. Microbial diversity tended to be lower T. cruzi-infected ones, in which Nocardiaceae and Pseudomonada-
in nymphs compared to male and female adult bugs, as indicated ceae were more abundant (Figure 8B). In bugs from haplogroup 3,
by Chao1 and Shannon diversity indices (Figure 7A and B, Staphylococcaceae, Comamonadaceae, Flavobacteriaceae, and Enter-
respectively), but this did not reach statistical significance obacteriaceae were more abundant in uninfected bugs, while
(F = 1.63, P = 0.21 and F = 1.56, P = 0.23, respectively). However, Pseudomonadaceae were more abundant in T. cruzi-infected bugs
at the level of beta diversity, there was a significant difference in (Figure 8C).

324
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

Figure 5. Analysis of Trypanosoma cruzi diversity in Triatoma dimidiata. (A) Maximum likelihood phylogenetic analysis of mini-exon sequences from T. dimidiata from Toledo
district with reference sequences from multiple DTUs, which indicated the clustering of Belize sequences with TcI and TcIV DTUs. (B) Maximum likelihood analysis of TcI
sequences from Belize. Sequences are color-coded by individual bugs, which carried an average of 10  5 mini-exon haplotypes per bug. Principal component analysis of this
TcI diversity indicated that there was no significant difference in mini-exon sequence diversity among T. dimidiata ITS-2 haplogroups (C) (PERMANOVA; F = 1.34, P = 0.21).
Significant differences in TcI parasite haplotypes were found among sex/developmental stages of the bugs (D) (PERMANOVA; F = 14.01, P = 0.0001). Significant differences
were found among villages (E) (PERMANOVA; F = 18.79, P = 0.011).

Discussion domestic transmission of the parasite. The role of dogs as a major


host in this transmission network is in agreement with observa-
The aim of this study was to better characterize T. cruzi tions in other regions (Cohen and Gürtler, 2001; Gurtler et al.,
transmission cycles in southern Belize, and the potential risk for 2007; Dumonteil et al., 2018; Flores-Ferrer et al., 2019), and
human infection, as limited information is available on these strengthens the idea of reducing infection in dogs as part of T. cruzi
aspects. The metabarcoding approach used provided a detailed control interventions (Reithinger et al., 2005; Gurtler et al., 2009).
snapshot of Chagas disease ecology and epidemiology in this Furthermore, the identification of blood meals taken on sylvatic
region. hosts in bugs collected inside/around houses highlights the strong
The presence of several T. dimidiata haplogroups in the region connectivity between domestic and sylvatic habitats through the
was first confirmed, in agreement with previous observations dispersal of bugs (Dumonteil et al., 2007; Polonio et al., 2009;
(Dorn et al., 2009). However, the epidemiological relevance of their Barbu et al., 2010) and/or the proximity of these hosts. Frequent
taxonomic distinction remains questionable, as the sympatry of host-switching provides opportunities for cross-species T. cruzi
these vectors seems to be associated with indistinguishable blood transmission among sylvatic and domestic hosts and eventually
feeding profiles, so that they are involved in a shared host-feeding humans. Thus the transmission network observed here in southern
network. The identification of humans as a major feeding host Belize is similar to that observed in the Yucatan, Mexico
further indicates a significant risk for infection, which contrasts (Dumonteil et al., 2018).
with the lack of current reporting of human cases by the Ministry Differences were observed in the feeding profile of nymphs and
of Health (2014). Several common domestic animal species were adults, with nymphs feeding on dogs rather than humans, while
found to be involved as feeding hosts and may play a role in adults fed more on humans than on dogs. Such differences may

325
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

Figure 6. Geographic diversity of Trypanosoma cruzi. Maximum clade credibility trees were constructed for TcIV (A) and TcI (B) sequences. Sequences are color-coded
according to country of origin. Uncertainty in time of divergence is indicated for major nodes. Note the clustering of sequences from Belize/Central America compared to
sequences from Mexico/USA.

reflect differences in mobility, as nymphs are unable to fly. Indeed, Genetic differences in TcI parasites were nonetheless detected
it has previously been found that sleeping habits in southern between nymphs and adults, which may reflect their different
Mexico could differentially affect host availability by nymphs and blood sources. The analysis of a larger sample size would help
adults (Waleckx et al., 2016), and nymphs may have easier access to clarify this point. Differences in TcI parasites among villages were
dogs compared to humans in southern Belize. also observed, suggesting the local differentiation of strains. This is
The analysis of T. cruzi diversity was in agreement with previous further supported by the analysis of strains from a broader
reports identifying TcI and TcIV in bugs from Cayo and Toledo geographic range, which indicated a remarkable differentiation of
districts (Dorn et al., 2017), as well as in T. dimidiata from both TcI and TcIV parasites from Toledo, Belize. Central American
Guatemala (Orantes et al., 2018). Other DTUs found in vectors from TcI strains clustered separately from North American, as well as
southern Mexico (Lopez-Cancino et al., 2015; Dumonteil et al., from Colombian and other South American strains. This differs
2018) were not detected so far in Belize, but due to the limited from a previous study in which North and Central American TcI
sample size, their presence at low frequency cannot be excluded. parasites clustered together (Dorn et al., 2017). This discrepancy is
Sequence analysis further indicated that identical or highly similar likely due to the use in the present study of the mini-exon marker
TcI parasites from Toledo district circulated among the distinct T. rather than the 18S, which provides increased resolution of within-
dimidiata ITS-2 haplogroups, strengthening the conclusion that DTU genetic diversity of T. cruzi (Majeau et al., 2019, 2021). While a
these haplogroups are involved in a shared parasite transmission subdivision of TcIV between North and South American clades had
cycle. Thus, ITS-2 haplogroups would play a similar role in the been proposed (Yeo et al., 2011), the present study results suggest
epidemiology of T. cruzi transmission to humans, as proposed further geographic substructuring of TcIV between Central and
before in the Yucatan Peninsula (Herrera-Aguilar et al., 2009). North America. The Maya Mountains in western Toledo (Figure 1),

326
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

Figure 7. Diversity of Triatoma dimidiata microbiota. Comparison of Chao1 (A) and Shannon (B) alpha diversity of the microbiota from males, females, and nymphs (F = 1.64,
P = 0.21 and F = 1.56, P = 0.22, respectively). (C) Beta diversity of microbiota composition according to sex/developmental stage. Numbers in parenthesis next to bars indicate
group sample size. (D) NMDS analysis of microbiota diversity according to sex/developmental stage (F = 1.82, P = 0.04).

which culminate at 1120 m above sea level, may serve as a natural host skin or other environmental contamination (Eichler and
barrier isolating southern Belize. As noted before, T. cruzi strain Schaub, 2002; Dumonteil et al., 2018; Rodriguez-Ruano et al.,
differentiation may interfere with serological diagnostic perfor- 2018). Thus, interfering with microbiota development may provide
mance (Majeau et al., 2021), and lead to variations in the clinical alternative strategies for vector control. In a similar manner, the
presentation and progression of Chagas disease (Zingales, 2017). study data also hint at possible differences in microbiota
The diverse microbiota identified in T. dimidiata from Belize composition between T. cruzi-infected and uninfected bugs, but
mirrored that found in bugs from Yucatan, Mexico (Dumonteil the limited sample size did not allow clear differences to be
et al., 2018), with Bacillales, Actinomycetales, Enterobacteriales, and established. Associations between T. cruzi infection and bacterial
Burkholderiales accounting for the majority of the microbiota. communities have been detected before in triatomines (Orantes
Nymphs presented a simplified microbial community, predomi- et al., 2018; Waltmann et al., 2019; Dumonteil et al., 2020a,b;
nated by only Bacillales and Actinomycetales, and multiple Murillo Solano et al., 2021), and may also be used to interfere with
additional families were found in adults. This contrasts with vectorial capacity. Thus, improving our understanding of the
previous observations that also found ontogenic changes in interactions between gut microbiota and T. cruzi in triatomines
microbiota in T. dimidiata from Colombia, in which nymphs had should be key for the development of transmission-blocking
a more diverse microbiota compared to adults (Murillo Solano interventions.
et al., 2021). Nonetheless, these ontogenic changes are consistent This study has some limitations, the major ones being the
with a maturation of the microbiota during nymphal development limited bug sample size and the biased sampling of bugs associated
through coprophagy, but also potentially through contacts with with human housing, so that it provides only a general view of

327
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

Conflict of interest

The authors declare no conflict of interest.

Acknowledgments

This work was funded in part by the Louisiana Board of


Regentsthrough the Board of Regents Support Fund (#LESASF
(2018-21)-RD-A-19) and grant #632083 from Tulane University
School of Public Health and Tropical Medicineto ED. JLD was a
recipient of a Ph.D. Fellowship from CONACyT, Mexico (No.457722)
and a PROMUV Fellowship from Universidad Veracruzana, Mexico
(PROMUV-2019,S16017347). The funders had no role in the study
design, data collection and analysis, decision to publish, or
preparation of the manuscript.

Appendix A. Supplementary data

Supplementary material related to this article can be found, in


the online version, at doi:https://doi.org/10.1016/j.ijid.2021.05.083.

References

Barbu C, Dumonteil E, Gourbière S. Characterization of the dispersal of non-


domiciliated Triatoma dimidiata through the selection of spatially explicit
models. PLoS Negl Trop Dis 2010;4:e777.
Belize Ministry of Health. Belize health sector strategic plan 2014–2024. Ministry of
Helath-PAHO; 2014 68p..
Bosseno MF, Barnabe C, Magallon Gastelum E, Lozano Kasten F, Ramsey J, Espinoza
B, et al. Predominance of Trypanosoma cruzi lineage I in Mexico. J Clin Microbiol
2002;40(2):627–32.
Bouckaert R, Vaughan TG, Barido-Sottani J, Duchene S, Fourment M, Gavryushkina
A, et al. BEAST 2.5: an advanced software platform for Bayesian evolutionary
analysis. PLoS Comput Biol 2019;15(4)e1006650.
Breniere SF, Waleckx E, Barnabe C. Over six thousand Trypanosoma cruzi strains
classified into discrete typing units (DTUs): attempt at an inventory. PLoS Negl
Figure 8. Changes in microbiota composition in Triatoma dimidiata. Microbiota Trop Dis 2016;10(8)e0004792.
composition is compared among ITS-2 haplogroups of adult T. dimidiata (A), and Buekens P, Cafferata ML, Alger J, Althabe F, Belizan JM, Bustamante N, et al.
between Trypanosoma cruzi-positive and uninfected bugs from ITS-2 haplogroup 2 Congenital transmission of Trypanosoma cruzi in Argentina, Honduras, and
(B) and haplogroup 3 (C). Numbers in parenthesis next to bars indicate the group Mexico: an observational prospective study. Am J Trop Med Hyg 2018;98
sample size. (2):478–85.
Cohen JE, Gürtler RE. Modeling household transmission of American trypanosomi-
asis. Science 2001;293:694–8.
Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y, et al. Ribosomal database
T. dimidiata ecology in the domestic habitat in southern Belize. project: data and tools for high throughput rRNA analysis. Nucleic Acids Res
These bug populations are, however, the most relevant to assess 2014;42(Database issue):D633–42.
Coura JR, Petana WB. American trypanosomiasis in British Honduras. II. The
the risk for human infection. On the other hand, multiple other
prevalence of Chagas’ disease in Cayo District. Ann Trop Med Parasitol 1967;61
factors, including differences in host availability among villages (3):244–50.
and habitats (intradomicile, peridomicile, and sylvatic), may lead Dhariwal A, Chong J, Habib S, King IL, Agellon LB, Xia J. Microbiome analyst: a web-
to local differences in T. dimidiata ecology, which could not be based tool for comprehensive statistical, visual and meta-analysis of micro-
biome data. Nucleic Acids Res 2017;45(W1):W180–8.
assessed here. This study nonetheless provides a valuable Dorn PL, Calderon C, Melgar S, Moguel B, Solorzano E, Dumonteil E, et al. Two
framework for the further exploration of these differences. distinct Triatoma dimidiata (Latreille, 1811) taxa are found in sympatry in
In conclusion, this study showed that T. dimidiata from multiple Guatemala and Mexico. PLoS Negl Trop Dis 2009;3(3):e393.
Dorn PL, McClure AG, Gallaspy MD, Waleckx E, Woods AS, Monroy MC, et al. The
ITS-2 haplogroups are involved in a complex network of feeding diversity of the Chagas parasite, Trypanosoma cruzi, infecting the main Central
host species in southern Belize, with extensive connections American vector, Triatoma dimidiata, from Mexico to Colombia. PLoS Negl Trop
between sylvatic and domestic hosts, and a significant risk for Dis 2017;11(9)e0005878.
Dumonteil E, Gourbière S, Barrera-Perez M, Rodriguez-Felix E, Ruiz-Piña H, Baños-
human infection. Thus, increased disease surveillance is needed, Lopez O, et al. Geographic distribution of Triatoma dimidiata and transmission
and house infestation by triatomines should be targeted for control dynamics of Trypanosoma cruzi in the Yucatan peninsula of Mexico. Am J Trop
(Waleckx et al., 2015a,b). Both TcI and TcIV parasites were Med Hyg 2002;67:176–83.
Dumonteil E, Tripet F, Ramirez-Sierra MJ, Payet V, Lanzaro G, Menu F. Assessment of
identified in these vectors, and sequence analysis showed the
Triatoma dimidiata dispersal in the Yucatan peninsula of Mexico using
local differentiation of these strains, which may interfere with morphometry and microsatellite markers. Am J Trop Med Hyg 2007;76:930–7.
serological diagnosis (Majeau et al., 2021) and lead to variable Dumonteil E, Ferral J, Euan-García M, Chavez-Nuñez L, Ramirez-Sierra MJ.
Usefullness of community participation for the fine-scale monitoring of non-
disease manifestations (Zingales, 2017). The analysis of triatomine
domiciliated triatomines. J Parasitol 2009;95(2):469–71.
microbiota further suggested that T. cruzi parasite interactions Dumonteil E, Nouvellet P, Rosecrans K, Ramirez-Sierra MJ, Gamboa-Leon R, Cruz-
with bacteria should be better characterized, as this may lead to Chan V, et al. Eco-bio-social determinants for house infestation by non-
novel transmission-blocking strategies to mitigate the impact of domiciliated Triatoma dimidiata in the Yucatan peninsula, Mexico. PLoS Negl
Trop Dis 2013;7(9):e2466.
Chagas disease. Dumonteil E, Ramirez-Sierra MJ, Pérez-Carrillo S, Teh-Poot C, Herrera C, Gourbière S,
et al. Detailed ecological associations of triatomines revealed by metabarcoding
Ethical approval based on next-generation sequencing: linking triatomine behavioral ecology
and Trypanosoma cruzi transmission cycles. Sci Rep 2018;8(1):4140.
Dumonteil E, Elmayan A, Majeau A, Tu W, Duhon B, Marx P, et al. Genetic diversity of
Not applicable. Trypanosoma cruzi parasites infecting dogs in southern Louisiana sheds light on

328
R. Polonio, J. López-Domínguez, C. Herrera et al. International Journal of Infectious Diseases 108 (2021) 320–329

parasite transmission cycles and serological diagnostic performance. PLoS Negl Murillo-Solano C, Lopez-Dominguez J, Gongora R, Rojas-Gulloso A, Usme-Ciro J,
Trop Dis 2020a;14(12)e0008932. Perdomo-Balaguera E, et al. Diversity and interactions among triatomine bugs,
Dumonteil E, Pronovost H, Bierman EF, Sanford A, Majeau A, Moore R, et al. Interactions their blood feeding sources, gut microbiota and Trypanosoma cruzi in the Sierra
among Triatoma sanguisuga blood feeding sources, gut microbiota and Trypano- Nevada de Santa Marta in Colombia. Sci Rep 2021;11(1):12306.
soma cruzi diversity in southern Louisiana. Mol Ecol 2020b;29(19):3747–61. Orantes LC, Monroy C, Dorn PL, Stevens L, Rizzo DM, Morrissey L, et al. Uncovering
Eichler S, Schaub GA. Development of symbionts in triatomine bugs and the effects vector, parasite, blood meal and microbiome patterns from mixed-DNA
of infections with trypanosomatids. Exp Parasitol 2002;100(1):17–27. specimens of the Chagas disease vector Triatoma dimidiata. PLoS Negl Trop
Flores-Ferrer A, Waleckx E, Rascalou G, Dumonteil E, Gourbière S. Trypanosoma cruzi Dis 2018;12(10)e0006730.
transmission dynamics in a synanthropic and domesticated host community. Petana WB. American trypanosomiasis in British Honduras X: natural habitats and
PLoS Negl Trop Dis 2019;13(12)e0007902. ecology of Triatoma dimidiata in the El Cayo and Toledo districts, and the
Gamboa-León R, Ramirez-Gonzalez C, Pacheco-Tucuch F, O’Shea M, Rosecrans K, prevalence of infection with Trypanosoma cruzi in the wild-caught bugs. Ann
Pippitt J, et al. Chagas disease among mothers and children in rural Mayan Trop Med Parasitol 1972;65:169–78.
communities. Am J Trop Med Hyg 2014;91(2):348–53. Polonio R, Ramirez-Sierra MJ, Dumonteil E. Dynamics and distribution of house
Garrison E, Marth G. Haplotype-based variant detection from short-read sequenc- infestation by Triatoma dimidiata in central and southern Belize. Vector Borne
ing. arXiv preprint 2012;1–9 (arXiv:1207.3907 [q-bio.GN]). Zoonotic Dis 2009;9(1):19–24.
Gurtler RE, Cecere MC, Lauricella MA, Cardinal MV, Kitron U, Cohen JE. Domestic Ramos-Ligonio A, Torres-Montero J, Lopez-Monteon A, Dumonteil E. Extensive
dogs and cats as sources of Trypanosoma cruzi infection in rural northwestern diversity of Trypanosoma cruzi discrete typing units circulating in Triatoma
Argentina. Parasitology 2007;134(Pt 1):69–82. dimidiata from central Veracruz, Mexico. Infect Genet Evol 2012;12(7):1341–3.
Gurtler RE, Ceballos LA, Stariolo R, Kitron U, Reithinger R. Effects of topical Reithinger R, Ceballos L, Stariolo R, Davies CR, Gurtler RE. Chagas disease control:
application of fipronil spot-on on dogs against the Chagas disease vector deltamethrin-treated collars reduce Triatoma infestans feeding success on dogs.
Triatoma infestans. Trans R Soc Trop Med Hyg 2009;103(3):298–304. Trans R Soc Trop Med Hyg 2005;99(7):502–8.
Hammer Ø, Harper DAT, Ryan PD. PAST: paleontological statistics software package Rodriguez-Ruano SM, Skochova V, Rego ROM, Schmidt JO, Roachell W, Hypsa V, et al.
for education and data analysis. Palaeontol Electron 2001;4(1):9. Microbiomes of North American Triatominae: the grounds for Chagas disease
Herrera C, Truyens C, Dumonteil E, Alger J, Sosa-Estani S, Cafferata ML, et al. epidemiology. Front Microbiol 2018;9:1167.
Phylogenetic analysis of Trypanosoma cruzi from pregnant women and Torres-Silva CF, Repoles BM, Ornelas HO, Macedo AM, Franco GR, Junho Pena SD,
newborns from Argentina, Honduras and Mexico suggests an association of et al. Assessment of genetic mutation frequency induced by oxidative stress in
parasite haplotypes with congenital transmission of the parasite. J Mol Diagn Trypanosoma cruzi. Genet Mol Biol 2018;41(2):466–74.
2019;21(6):1095–105. Villanueva-Lizama L, Teh-Poot C, Majeau A, Herrera C, Dumonteil E. Molecular
Herrera-Aguilar M, Be-Barragán LA, Ramirez-Sierra MJ, Tripet F, Dorn P, Dumonteil genotyping of Trypanosoma cruzi by next-generation sequencing of the mini-
E. Identification of a large hybrid zone between sympatric sibling species of exon gene reveals infections with multiple parasite DTUs in Chagasic patients
Triatoma dimidiata in the Yucatan peninsula, Mexico, and its epidemiological from Yucatan, Mexico. J Inf Dis 2019;219(12):1980–8.
importance. Infect Genet Evol 2009;9:1345–51. Waleckx E, Camara-Mejia J, Ramirez-Sierra MJ, Cruz-Chan V, Rosado-Vallado M,
Hotez PJ, Bottazzi ME, Franco-Paredes C, Ault SK, Periago MR. The neglected tropical Vazquez-Narvaez S, et al. An innovative ecohealth intervention for Chagas
diseases of Latin America and the Caribbean: a review of disease burden and disease vector control in Yucatan, Mexico. Trans R Soc Trop Med Hyg 2015a;109
distribution and a roadmap for control and elimination. PLoS Negl Trop Dis (2):143–9.
2008;2(9):e300. Waleckx E, Gourbière S, Dumonteil E. Intrusive triatomines and the challenge of
Hotez PJ, Dumonteil E, Betancourt Cravioto M, Bottazzi ME, Tapia-Conyer R, adapting vector control practices. Mem Inst Oswaldo Cruz 2015b;110(3):
Meymandi S, et al. An unfolding tragedy of Chagas disease in North America. 324–38.
PLoS Negl Trop Dis 2013;7(10):e2300. Waleckx E, Pasos-Alquicira R, Ramirez-Sierra MJ, Dumonteil E. Sleeping habits affect
Jaramillo R, Bryan JP, Schur J, Pan AA. Prevalence of antibody to Trypanosoma cruzi in access to host by Chagas disease vector Triatoma dimidiata. Parasit Vectors
three populations in Belize. Am J Trop Med Hyg 1997;57:298–301. 2016;9(1):568.
Justi SA, Cahan S, Stevens L, Monroy C, Lima-Cordon R, Dorn PL. Vectors of diversity: Waltmann A, Willcox AC, Balasubramanian S, Borrini Mayori K, Mendoza Guerrero
genome wide diversity across the geographic range of the Chagas disease vector S, Salazar Sanchez RS, et al. Hindgut microbiota in laboratory-reared and wild
Triatoma dimidiata sensu lato (Hemiptera: Reduviidae). Mol Phylogenet Evol Triatoma infestans. PLoS Negl Trop Dis 2019;13(5)e0007383.
2018;120:144–50. Yeo M, Mauricio IL, Messenger LA, Lewis MD, Llewellyn MS, Acosta N, et al.
Lee BY, Bacon KM, Bottazzi ME, Hotez PJ. Global economic burden of Chagas disease: Multilocus sequence typing (MLST) for lineage assignment and high resolution
a computational simulation model. Lancet Infect Dis 2013;13(4):342–8. diversity studies in Trypanosoma cruzi. PLoS Negl Trop Dis 2011;5(6):e1049.
Llewellyn MS, Miles MA, Carrasco HJ, Lewis MD, Yeo M, Vargas J, et al. Genome-scale Zingales B. Trypanosoma cruzi genetic diversity: something new for something
multilocus microsatellite typing of Trypanosoma cruzi discrete typing unit I known about Chagas disease manifestations, serodiagnosis and drug sensitivity.
reveals phylogeographic structure and specific genotypes linked to human Acta Trop 2017;184:38–52.
infection. PLoS Pathog 2009;5(5)e1000410. Zingales B, Andrade SG, Briones MR, Campbell DA, Chiari E, Fernandes O, et al. A new
Lopez-Cancino SA, Tun-Ku E, Felix HK, Ibarra-Cerdena CN, Izeta-Alberdi A, Pech- consensus for Trypanosoma cruzi intraspecific nomenclature: second revision
May A, et al. Landscape ecology of Trypanosoma cruzi in the southern Yucatan meeting recommends TcI to TcVI. Mem Inst Oswaldo Cruz 2009;104(7):1051–4.
Peninsula. Acta Trop 2015;151:58–72. Zingales B, Miles MA, Campbell DA, Tibayrenc M, Macedo AM, Teixeira MM, et al.
Majeau A, Herrera C, Dumonteil E. An improved approach to Trypanosoma cruzi The revised Trypanosoma cruzi subspecific nomenclature: rationale, epidemio-
molecular genotyping by next-generation sequencing of the mini-exon gene. logical relevance and research applications. Infect Genet Evol 2012;12(2):
Methods Mol Biol 2019;1955:47–60. 240–53.
Majeau A, Murphy L, Herrera C, Dumonteil E. Assessing Trypanosoma cruzi parasite
diversity through comparative genomics: implications for disease epidemiolo-
gy and diagnostics. Pathogens 2021;10:212.

329

View publication stats

You might also like