Hostname: page-component-745bb68f8f-b6zl4 Total loading time: 0 Render date: 2025-02-11T01:41:22.905Z Has data issue: false hasContentIssue false

A systematic review of the Trypanosoma cruzi genetic heterogeneity, host immune response and genetic factors as plausible drivers of chronic chagasic cardiomyopathy

Published online by Cambridge University Press:  13 September 2018

Paula Jiménez
Affiliation:
Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
Jesús Jaimes
Affiliation:
Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
Cristina Poveda
Affiliation:
Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, USA
Juan David Ramírez*
Affiliation:
Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
*
Author for correspondence: Juan David Ramírez, E-mail: juand.ramirez@urosario.edu.co

Abstract

Chagas disease is a complex tropical pathology caused by the kinetoplastid Trypanosoma cruzi. This parasite displays massive genetic diversity and has been classified by international consensus in at least six Discrete Typing Units (DTUs) that are broadly distributed in the American continent. The main clinical manifestation of the disease is the chronic chagasic cardiomyopathy (CCC) that is lethal in the infected individuals. However, one intriguing feature is that only 30–40% of the infected individuals will develop CCC. Some authors have suggested that the immune response, host genetic factors, virulence factors and even the massive genetic heterogeneity of T. cruzi are responsible of this clinical pattern. To date, no conclusive data support the reason why a few percentages of the infected individuals will develop CCC. Therefore, we decided to conduct a systematic review analysing the host genetic factors, immune response, cytokine production, virulence factors and the plausible association of the parasite DTUs and CCC. The epidemiological and clinical implications are herein discussed.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2018 

Introduction

Chagas disease, also known as American trypanosomiasis, is caused by the parasite Trypanosoma cruzi. It is a complex zoonosis that is widely distributed throughout the continent of America. The infection can be acquired by contact with infected triatomine faeces, blood transfusion, laboratory accidents, or by oral and congenital transmission. Chagas disease is a major public-health problem, with approximately 6 million people infected in South America and 25 million at risk of infection, according to the statistics published by the World Health Organization (WHO, 2017). The immigration of infected people from endemic countries has also recently led to Chagas disease becoming an important health issue in other continents, such as Europe, the United States, Australia, Asia and Africa (Rassi et al., Reference Rassi, Rassi and Marin-Neto2009; Kuete, Reference Kuete2013; Jackson et al., Reference Jackson, Pinto and Pett2014; Strasen et al., Reference Strasen, Williams, Ertl, Ritter, Zoller and Stich2014; Angheben et al., Reference Angheben, Buonfrate, Gobbi, Bisoffi, Boix, Pupella and Gandini2015). Chagas disease comprises two stages: an acute phase that occurs about 1 week after the initial infection, and a chronic phase that affects about 30–40% of infected patients for which cardiomyopathy is the most frequent and severe clinical manifestation (Rassi et al., Reference Rassi, Rassi and Marin-Neto2009; Bern et al., Reference Bern, Kjos, Yabseley and Montgomery2011; Dias et al., Reference Dias, Ramos, Gontijo, Luquetti, Shikanai-Yasuda, Coura, Torres, Melo, Almeida, Oliveira, Silveira, Rezende, Pinto, Ferreira, Rassi, Fragata, Sousa, Correia, Jansen, Andrade, Britto, Pinto, Rassi, Campos, Abad-Franch, Santos, Chiari, Hasslocher-Moreno, Moreira, Marques, Silva, Marin-Neto, Galvão, Xavier, Valente, Carvalho, Cardoso, Silva, Costa, Vivaldini, Oliveira, Valente, Lima and Alves2016).

Understanding the epidemiology of Chagas disease, which is considered an anthropozoonosis, is complex. One of the pivotal factors that makes the epidemiological information and the variation in clinical manifestations even more complex is the intraspecific genetic diversity among T. cruzi taxon, for which different methods of classification have been developed based on new identification techniques (Zingales et al., Reference Zingales, Miles, Campbell, Tibayrenc, Macedo, Teixeira, Schijman, Llewellyn, Lages-Silva, Machado, Andrade and Sturm2012; Lima et al., Reference Lima, Espinosa-Álvarez, Ortiz, Camargo, Teixeira, Trejo-Varón, Carranza, Pinto, Serrano and Buck2015a; Brenière et al., Reference Brenière, Barnabé and Waleckx2016). There are seven subtypes within this classification, known as discrete typing units (DTUs), which include T. cruzi I (TcI, which includes TcIDom), T. cruzi II (TcII), T. cruzi III (TcIII), T. cruzi IV (TcIV), T. cruzi V (TcV) and T. cruzi VI (TcVI), and the recently described genotype associated with anthropogenic bats designated TcBat (Pinto et al., Reference Pinto, Kalko, Cottontail, Cottontail and Wellinghausen2012; Guhl et al., Reference Guhl, Auderheide and Ramírez2014; Hernández et al., Reference Hernández, Cucunubá, Parra, Toro, Zambrano and Ramírez2014; Ramírez et al., Reference Ramírez, Hernández, Cucunubá, Montilla, Flórez, Zambrano and Parra2014; Lima et al., Reference Lima, Espinosa-Álvarez, Ortiz, Camargo, Teixeira, Trejo-Varón, Carranza, Pinto, Serrano and Buck2015a, Reference Lima, Espinosa-Álvarez, Pinto, Cavazzana, Pavan, Carranza, Lim, Campaner, Takata, Camargo, Hamilton and Teixeira2015b).

Around 30–40% of the infected patients in the symptomatic chronic phase exhibit one or more of the following cardiac clinical conditions: tachycardia, bradyarrhythmias, apical aneurysms, cardiac failure (as a late manifestation with its typical signs and symptoms but a higher mortality rate when caused by Chagas disease), thromboembolic phenomena and sudden death, the latter being the leading cause of death from Chagas cardiac disease, more commonly displayed in the early stages (even in asymptomatic patients and is associated with ventricular fibrillation or tachycardia and less frequently to atrioventricular block or sinus node disease) (Rassi et al., Reference Rassi, Rassi and Marin-Neto2010, Reference Rassi, Rassi and Marcondes de Rezende2012, Reference Rassi, Rassi and Marin-Neto2015). Electrocardiographic manifestations such as premature ventricular beats, ST-T segment changes, abnormal Q waves, voltage drop in the QRS complex, right bundle branch block and left anterior fascicular block are frequent, with the last two symptoms commonly occurring at the same time. Monitoring with a Holter device usually reveals ventricular compromise including ventricular dysfunction and tachycardia (Rassi et al., Reference Rassi, Rassi and Marin-Neto2015; Sánchez-Montalvá et al., Reference Sánchez-Montalvá, Salvador, Rodríguez-Palomares, Sulleiro, Sao-Avilés, Roure, Valerio, Evangelista and Molina2016). Chronic chagasic cardiomyopathy (CCC) is considered a multifactorial event in infected patients, with the immune system, host factors and the genetic diversity of the parasite all playing fundamental roles.

Several studies have been conducted over the past 40 years to elucidate the genetic variation of T. cruzi across its geographical distribution and the associations with host and vector species. Also, different reviews have been published in the literature showing the plausible effect of T. cruzi heterogeneity in the disease outcome and clinical features of patients infected with this parasite (Zingales et al., Reference Zingales, Andrade, Briones, Campbell, Chiari, Fernandes, Guhl, Lages-Silva, Macedo, Machado, Miles, Romanha, Sturm, Tibayrenc and Schijman2009; Zingales et al., Reference Zingales, Miles, Campbell, Tibayrenc, Macedo, Teixeira, Schijman, Llewellyn, Lages-Silva, Machado, Andrade and Sturm2012; Messenger et al., Reference Messenger, Ramirez, Llewellyn, Guhl and Miles2016; Zingales, Reference Zingales2017). Different studies suggest that this variation may or may not be associated with the different clinical manifestations of Chagas disease. Since 1981, the role of T. cruzi genetic diversity on the pathogenicity of Chagas disease has been investigated by Miles et al., with no conclusive answer so far (Miles et al., Reference Miles, Póvoa, Prata, Cedillos, De Souza and Macedo1981). Therefore, we conducted a systematic review of cutting-edge research in this field to elucidate the possible associations between T. cruzi genetic heterogeneity, host immune response and genetic factors, and the clinical manifestations of CCC considering for the first time, the three players in disease outcome (parasite genetic variability, host genetics and host immune response).

Molecular epidemiology of Chagas disease

The T. cruzi parasite comprises a heterogeneous population that displays clonal and/or sexual propagation due to the different cycles of transmission, and the possibility of genetic exchange that can be found in nature and has been previously reported in vitro (Gaunt et al., Reference Gaunt, Yeo, Frame, Stothard, Carrasco, Taylor, Mena, Solis, Miles, Acosta, de Arias and Miles2003; Sturm et al., Reference Sturm, Vargas, Westenberger, Zingales and Campbell2003; Westenberger et al., Reference Westenberger, Campbell, Sturm and Barnabé2005; Llewellyn et al., Reference Llewellyn, Miles, Carrasco, Lewis, Yeo, Vargas, Torrico, Diosque, Valente, Valente and Gaunt2009; Ramírez et al., Reference Ramírez, Guhl, Messenger, Lewis, Montilla, Cucunuba and Llewellyn2012; Ramírez and Llewellyn, Reference Ramírez and Llewellyn2014; Brenière et al., Reference Brenière, Barnabé and Waleckx2016). T. cruzi is genetically diverse and is classified into at least six DTUs known as TcI–TcVI based on different molecular markers and biological features. TcI and TcII are considered as two pure lines with a point of evolutionary separation of approximately 1–3 million years ago. Some authors consider TcIII and TcIV to be the result of hybridization between TcI and TcII, whereas TcV and TcVI are generally accepted to be the result of hybridization between TcII and TcIII (Gaunt et al., Reference Gaunt, Yeo, Frame, Stothard, Carrasco, Taylor, Mena, Solis, Miles, Acosta, de Arias and Miles2003; Sturm et al., Reference Sturm, Vargas, Westenberger, Zingales and Campbell2003; Westenberger et al., Reference Westenberger, Campbell, Sturm and Barnabé2005; Zingales et al., Reference Zingales, Miles, Campbell, Tibayrenc, Macedo, Teixeira, Schijman, Llewellyn, Lages-Silva, Machado, Andrade and Sturm2012; Ramírez et al., Reference Ramírez, Hernández, Cucunubá, Montilla, Flórez, Zambrano and Parra2014; Brenière et al., Reference Brenière, Barnabé and Waleckx2016; Zingales, Reference Zingales2017). Recently, other authors have proposed divergent evolutionary pathways suggesting three different ancestors (Brenière et al., Reference Brenière, Barnabé and Waleckx2016). Future studies are needed to fully understand the emergence of T. cruzi DTUs. Over the last decade, two emergent genotypes have been reported; one associated with anthropogenic bats in Brazil, Colombia, Ecuador and Panama and designated TcBat (Ramírez et al., Reference Ramírez, Hernández, Cucunubá, Montilla, Flórez, Zambrano and Parra2014; Pinto et al., Reference Pinto, Ocaña-Mayorga, Tapia, Lobos, Zurita, Aguirre-Villacís, MacDonals, Villacis, Lima, Teixeira, Grijalva and Perkins2015), and a clonal genotype linked to human infections within TcI designated TcIDom (Llewellyn et al., Reference Llewellyn, Miles, Carrasco, Lewis, Yeo, Vargas, Torrico, Diosque, Valente, Valente and Gaunt2009; Ramírez et al., Reference Ramírez, Guhl, Messenger, Lewis, Montilla, Cucunuba and Llewellyn2012; Zumaya-Estrada et al., Reference Zumaya-Estrada, Messenger, Lopez-Ordonez, Lewis, Flores-Lopez, Martínez-Ibarra, Pennington, Cordon-Rosales, Carrasco, Segovia, Miles and Llewellyn2012; Sánchez and Ramírez, Reference Sánchez and Ramírez2013; Ramírez and Hernández, Reference Ramírez and Hernández2017). However, these emergent genotypes are not as yet considered true DTUs by international consensus.

The DTUs of T. cruzi are broadly distributed across the American continent in diverse ecotopes (domestic, peridomestic and sylvatic transmission cycles). Discrimination of the six DTUs has become an important issue in the molecular epidemiology of T. cruzi. There are many reports showing algorithms for the molecular characterization of these DTUs based on RAPDs, polymerase chain reaction (PCR)-RFLPs, qPCR, MLST, MLMT and DNA sequencing techniques, but to date there is no gold standard protocol that is accepted internationally for strain typing (Rozas et al., Reference Rozas, Doncker, Adaui, Coronado, Barnabé, Tibyarenc and Dujardin2007; Duffy et al., Reference Duffy, Bisio, Altcheh, Burgos, Diez, Levin, Favaloro, Freilij and Schijman2009; Lewis et al., Reference Lewis, Yeo, Llewellyn, Miles, Ma and Carrasco2009; Llewellyn et al., Reference Llewellyn, Miles, Carrasco, Lewis, Yeo, Vargas, Torrico, Diosque, Valente, Valente and Gaunt2009; Burgos et al., Reference Burgos, Diez, Vigliano, Bisio, Risso, Duffy, Cura, Brusses, Favaloro, Leguizamon, Lucero, Laguens, Levin, Favaloro and Schijman2010; Ramírez et al., Reference Ramírez, Guhl, Rendón, Rosas, Marin-Neto and Morillo2010; Yeo et al., Reference Yeo, Mauricio, Messenger, Lewis, Llewellyn, Acosta, Bhattacharyya, Diosque, Carrasco and Miles2011; Higuera et al., Reference Higuera, Guhl and Ramírez2013; Cura et al., Reference Cura, Duffy, Lucero, Bisio, Péneau, Jimenez-Coello, Calabuig, Gimenez, Valencia, Kjos, Santalla, Mahaney, Cayo, Nagel, Barcán, Málaga Machaca, Acosta, Brutus, Ocampo and Aznar2015). One of the most frequently used and reliable algorithms for T. cruzi typing has been reported by the research of Burgos and colleagues, Higuera and colleagues and Ramírez and colleagues, and has also been applied to the testing of clinical samples (mainly blood samples) (Burgos et al., Reference Burgos, Diez, Vigliano, Bisio, Risso, Duffy, Cura, Brusses, Favaloro, Leguizamon, Lucero, Laguens, Levin, Favaloro and Schijman2010; Ramírez et al., Reference Ramírez, Guhl, Rendón, Rosas, Marin-Neto and Morillo2010; Higuera et al., Reference Higuera, Guhl and Ramírez2013) (Fig. 1).

Fig. 1. Algorithm for DTU discrimination of T. cruzi.

Regarding the geographical distribution of T. cruzi DTUs, it is clear that TcI predominates in the terrestrial area from the south of North America to the north of Argentina and Chile, usually due to a sylvatic transmission cycle, but TcI is also a major cause of the disease in countries endemic for trypanosomiasis due to a domestic transmission cycle. By contrast, TcII, TcV and TcVI mainly occur in the southern cone, but this distribution may not be completely accurate since the area includes countries such as Bolivia for which data may be incomplete. TcIII and TcIV are generally characterized by their sylvatic transmission cycles in areas with tropical forest ecosystems (Carrasco et al., Reference Carrasco, Segovia, Llewellyn, Morocoima, Urdaneta-Morales, Martínez, Garcia, Rodríguez, Espinosa, de Noya, Díaz-Bello, Herrera, Fitzpatrick, Yeo, Miles and Feliciangeli2012; Guhl and Ramírez, Reference Guhl and Ramírez2013; Brenière et al., Reference Brenière, Barnabé and Waleckx2016). This is particularly illuminating given that there are distinct clinical differences between patients presenting with Chagas disease in these geographical regions. Strains appear to differ in terms of pathogenicity and the response to treatment. Both TcI and TcII–VI are associated with cardiac lesions in human infections, but it seems that only TcII, TcV and TcIV are associated with digestive tract lesions (Prata, Reference Prata2001), despite a report of digestive tract lesions in Colombia caused by TcI (Prata, Reference Prata2001). In general, however, TcI is considered to be less pathogenic with lower parasitaemias (Burgos et al., Reference Burgos, Bisio, Duffy, Levin, Schijman, Altcheh, Burgos Freilij, Valadares, Freitas, Macedo, Seidenstein, Macchi and Piccinali2007) and more chronic cases being asymptomatic compared with Chagas cases caused by TcII, TcV and TcVI in Argentina, Brazil, Chile, Paraguay and Uruguay (Luquetti et al., Reference Luquetti, Tavares, Siriano, Oliveira, Campos, Morais and Oliveira2015).

Additionally, some proteomic approaches have been conducted demonstrating differential protein expression among DTUs isolates (Telleria et al., Reference Telleria, Biron, Brizard, Demettre, Séveno, Barnabé, Ayala and Tibayrenc2010; Díaz et al., Reference Díaz, Torres and González2011). Moreover, there is an observed general partitioning of TcII–TcVI between sylvatic and domestic transmission cycles; with human disease cases associated with TcII, TcV and TcVI and TcIII, TcIV being predominantly sylvatic (Yeo et al., Reference Yeo, Acosta, Llewellyn, Sánchez, Adamson, Miles, López, González, Patterson, Gaunt, de Arias and Miles2005; Zingales et al., Reference Zingales, Miles, Campbell, Tibayrenc, Macedo, Teixeira, Schijman, Llewellyn, Lages-Silva, Machado, Andrade and Sturm2012; Messenger et al., Reference Messenger, Ramirez, Llewellyn, Guhl and Miles2016). However, according to the relationship between the recent TcBat isolates and anthropogenic deaths reported in countries such as Colombia and Brazil, the zoonotic potential of this genotype is being considered (Ramírez et al., Reference Ramírez, Hernández, Cucunubá, Montilla, Flórez, Zambrano and Parra2014). In addition, TcII has also been isolated from the same type of bats from which TcBat originated, and TcII has been shown phylogenetically to be closely related to a common ancestor of the TcI genotype (Lima et al., Reference Lima, Espinosa-Álvarez, Pinto, Cavazzana, Pavan, Carranza, Lim, Campaner, Takata, Camargo, Hamilton and Teixeira2015b). Recent advances in next generation sequencing technologies have allowed researchers to obtain three complete T. cruzi genomes. The first strain to be fully sequenced was CL Brener (TcVI), which revealed a high degree of repetitive elements along the core genome (El-Sayed et al., Reference El-Sayed, Bartholomeu, Ghedin, Delcher, Blandin, Westenberger, Caler, Cerqueira, Haas, Crabtree, Feldblyum, Hou, Koo, Lacerda, Pai, Pop, Salzberg, Shetty, Simpson, Van Aken, Wortman, White, Fraser, Myler, Aggarwal, Worthey, Anupama, Attipoe, Cadag, Fazelina, Huang, Louie, Nelson, Parsons, Pentony, Rinta, Robertson, Seyler, Sisk, Vogt, Stuart, Nilsson, Tran, Branche, Arner, Bontempi, Darban, Edwards, Ferella, Kindlund, Kluge, McKenna, Mizuno, Ochaya, Tammi, Andersson, Campbell, Machado, Teixeira, Åslund, Pettersson, Bringaud, Burton, McCulloch, Mottram, Ward, Carrington, Sharma, Da Silveira, De Jong, Osoegawa, Englund, Frasch, Sanchez, Gull, Wickstead, Horn, Klingbeil, Levin, Lorenzi, Ramirez and Tarleton2005). Similarly, the recently sequenced Esmeraldo (TcII) and Sylvio X10 (TcI) genomes revealed the association between repetitive elements and mucin-like proteins that are closely associated with parasite cell invasion and survival, (Franzén et al., Reference Franzén, Ochaya, Sherwood, Andersson, Lewis, Llewellyn and Miles2011) offering new insight into the relationship between T. cruzi DTUs and pathogenicity (Andersson, Reference Andersson2011).

Pathophysiology of CCC

Chagas disease is currently considered a multifactorial disease of infectious origin, since several factors such as the mechanisms of action by which the parasite invades, the virulence factors of the parasite and the polymorphic factors of the host come into play, which together determine the severity of the clinical manifestations of the disease (Prata, Reference Prata2001). Below, the pathophysiological factors involved in the different presentations of the disease will be discussed, as well as a brief description of the manifestations involved.

The acute phase of T. cruzi infection is mediated by both innate and acquired host immunity. Initially, the infection is mediated by phagocytosis with highly virulent metacyclic trypomastigotes being ingested by macrophages. Although most are destroyed within phagocytic vacuoles, some amastigotes escape this immunological action and initiate intracellular replication, causing cell damage and rupture, allowing the parasite to spread through the bloodstream. This leads to the activation of a cascade of cytokines, cationic proteins, complement and transferrin proteins, with T. cruzi inducing the expression of interferons and cytokines such as interleukin (IL)-10 and tumour necrosis factor (TNF)-α (Sanjabi et al., Reference Sanjabi, Zenewicz, Kamanaka and Flavell2009; Teixeira et al., Reference Teixeira, Hecht, Guimaro, Sousa and Nitz2011). Regarding acquired immunity, certain humoral factors play an important role in control of the infection, among which immunoglobulin G (IgG)2b antibodies are pivotal, which also mediate inflammatory activity during the acute phase. The activation of immune cells, such as CD4+ and CD8+ cells, also plays an important role in acquired immunity (Teixeira et al., Reference Teixeira, Hecht, Guimaro, Sousa and Nitz2011).

At the moment the parasite enters the bloodstream, it is directed to the sites with the highest predilection, which in this case are the central nervous system, the cardiac and oesophageal regions, and the colon muscle tissue, as these are sites where the parasite can most readily traverse the vascular endothelium and initiate cellular parasitism (Rassi et al., Reference Rassi, Marin Neto and Rassi2017). The initial process of transmigration through the vascular endothelium prior to transport of the parasite to its target tissues, has been reported by in vitro studies in human cells, and is facilitated by chemokines such as bradykinin and chemokine ligand 2 (CCL2). However, the molecules through which the parasite interacts with the endothelium are yet to be determined (Coates et al., Reference Coates, Sullivan, Makanji, Du, Olson, Muller, Engman and Epting2013).

On reaching and invading its target organs within the host, the parasite undergoes active multiplication during a sustained immunological reaction by the cells that involves CD4+ T lymphocytes, CD8+ and B lymphocytes. This cellular reaction leads to direct induction of anti-trypanosome cytotoxicity, secretion of cytokines and the production of antibodies against the parasite. Inflammatory myocardial lesions of both chronically infected animal and human models are predominantly composed of CD8+ rather than Th1 CD4+ lymphocytes, and increased expression of genes responsible for the production of proinflammatory cytokines and chemokines (especially interferon (IFN)-γ and transforming growth factor (TGF)-β±) has been observed. Other investigators have reported reduced production of regulatory T cells and their cytokines, such as IL-10 and IL-17 (Reis et al., Reference Reis, Jones, Tostes, Lopes, Gazzinelli, Colley and McCurley1993; Sanjabi et al., Reference Sanjabi, Zenewicz, Kamanaka and Flavell2009; Guedes et al., Reference Guedes, Gutierrez, Silva, Dellalibera-Joviliano, Rodrigues, Bendhack, Rassi, Schmidt, Maciel, Marin- Neto and Silva2012; Cupello et al., Reference Cupello, Souza, Nogueira, Laranja, Sabino, Coelho, Oliveira and Paes2014). These findings are consistent with an imbalance demonstrated by the upregulation of Th1 cells and the downregulation of Treg cell activity (Fig. 2).

Fig. 2. Cytokine profile during T. cruzi infection. The acute phase shows the immune system response and the chronic phase shows the activation of cytokines.

A study using mice susceptible (BALB/c) and non-susceptible (C57BL/6) to T. cruzi infection was performed to compare the expression of CD4+ T cells in both the acute and chronic phases of the disease. During the chronic phase, a T. cruzi-specific product was amplified by PCR at higher quantities in the surviving BALB/c mice than in the C57BL/6 mice, suggesting that a greater number of parasites were present in the susceptible mice during this phase. Immunological analysis of the chronic phase samples revealed greater expression of Th1 and inflammatory cytokines, such as IFN-γ, TNF and IL-2, which triggered the low but significant activation of CD4+ T cells in the BALB/c mice. However, this cell type was not detected in the infected C57BL/6 mice, in which low levels of proinflammatory cytokines (IFN-γ and IL-2) were detected. Similarly, in BALB/c mice, the production of IL-10 and TGF-β increased, which was not observed in C57BL/6 mice. These results indicated that mice which survive the acute phase and then enter the chronic phase with parasites present in their cardiac tissue trigger a response associated with Th1 cells, but not a Th17 response (Sanoja et al., Reference Sanoja, Carbajosa, Fresno and Gironès2013).

It is also necessary to emphasize the importance of the Toll-like receptors (TLR) since they are responsible for immune recognition of parasites via pathogen-associated molecular patterns. T. cruzi has chemical structures that stimulate specific TLRs, which subsequently induce the production of nitric oxide and proinflammatory cytokines by monocytic cells. Among them, TLR-2 recognizes trypomastigote-derived glycosylphosphatidylinositol (tGPI) anchored in the mucin-like glycoproteins, TLR-4 recognizes the epimastigote glycoinositolphospholipid (eGIPL) that induces nuclear factor (NF)-κB, TLR-7 recognizes the parasitic RNA and TLR-9 recognizes DNA with abundant oligodeoxynucleotide unmethylated CpG motifs stimulating the cytochemical responses of both macrophages and dendritic cells (de Souza et al., Reference de Souza, de Carvalho and Barrias2010; Rodrigues et al., Reference Rodrigues, Oliveira and Bellio2012).

Other factors related to cardiac involvement are neurological or microvascular alterations, immune-mediated tissue injury and parasite-dependent damage. These play a secondary role in the development of cardiac lesions and complications. However, some authors consider these critical factors in the persistence of parasites and in the inflammatory reaction and the initiation and progression of chronic myocarditis (Tarleton and Zhang, Reference Tarleton and Zhang1999, Marin-Neto et al., Reference Marin-Neto, Rassi and Maciel2015). Immune-mediated tissue injury is caused by polymorphonuclear leucocyte infiltration and the production of deleterious cytokines, mechanisms that are probably triggered by the persistence of the parasite in the tissue. Autoimmunity generated by molecular mimicry with parasite antigens and the resulting polyclonal activation have been reported; however, validation of autoimmunity is difficult and therefore remains a controversial issue (Tarleton, Reference Tarleton1991, Reference Tarleton2003). Evidence for an additional potential autoimmune mechanism came from the detection of mitochondrial DNA from the parasite in the genome of chickens in a model in which infection was induced in the egg phase (Teixeira et al., Reference Teixeira, Hecht, Guimaro, Sousa and Nitz2011).

Metabolomic studies have revealed that the proteasome is the key protease in the generation of peptides for the presentation of antigens through MHC-I. In one study, it was shown that the biosynthesis of the immunoproteasome subunits Bl1, B2i and B5i, as well as PA28B, TAP1 and MCH-I (in macrophages via the SAPK/JNK signalling pathway) were downregulated in HeLa cells by T. cruzi, although the last three were not degraded by the parasite (Camargo et al., Reference Camargo, Faria, Kloss, Favali, Kuckelkorn, Kloetzel, de Sá and Lima2014). By contrast, the parasite does not affect the transcription or expression of proteins of the constitutive subunits of the proteasome. This provides evidence that this protozoan is able to modulate infection through post-transcriptional mechanisms that affect the translation of host proteins, as well as limiting their recognition by CD8+ cells, thus favouring parasite evasion from the immune response (Camargo et al., Reference Camargo, Faria, Kloss, Favali, Kuckelkorn, Kloetzel, de Sá and Lima2014; Tarleton, Reference Tarleton2015).

Thus, when the protozoan comes into contact with neurons and glial cells, it carries out the process of cell invasion by binding to receptor tyrosine kinase A and C (TrkA and TrkC), which are normally activated by neurotrophin (NT) nerve growth factor and NT-3, respectively, and whose activation is essential for maintenance of the nervous system (Chuenkova and Pereira, Reference Chuenkova and PereiraPerrin2009; Aridgides et al., Reference Aridgides, Salvador and PereiraPerrin2013). Recognition of these receptors occurs through parasite-derived neuropathic factor (PDNF), a GPI-linked neuraminidase/trans-sialidase (TS), whose interaction, as with other protein kinases (Akt), is independent of sialic acid (Chuenkova and Pereira, Reference Chuenkova and PereiraPerrin2011). Subsequently, these same receptors were identified in cardiomyocytes (Meloni et al., Reference Meloni, Caporali, Graiani, Lagrasta, Katare, Van Linthout, Spillmann, Campesi, Madeddu, Quaini and Emanueli2010; Aridgides et al., Reference Aridgides, Salvador and PereiraPerrin2013), where it was shown that these cells are also invaded via a mechanism that involves PDNF binding to TrkC receptor primarily, or TrkA receptor, which offers some protection against oxidative stress and parasite invasion (Aridgides et al., Reference Aridgides, Salvador and PereiraPerrin2013).

Parasite invasion induces multiple responses in the heart. Studies have shown that Cox2 mRNA and protein expression is induced in murine models during parasitic loading and myocarditis. This upregulation is also associated with the induction of cardiac dysfunction markers such as endothelin-1 (ET-1) and atrial natriuretic peptide (ANP), the first considered important in signalling pathways that allow Ca activation of ERK1/2 leading to cyclin-D1 expression and inflammation-related gene expression. In addition, Cox2 is induced via the Ca/calcineurin/NFAT pathway allowing overproduction of eicosanoids (thromboxane A2 (TXA2) and prostaglandins E2 and F2a) (Petkova et al., Reference Petkova, Tanowitz, Magazine, Factor, Chan, Pestell, Bouzahzah, Douglas, Shtutin, Morris, Tsang, Weiss, Christ, Wittner and Huang2000; Salomone et al., Reference Salomone, Caeiro, Madoery, Amuchastegui, Omelinauk, Juri and Kaski2001; Hassan et al., Reference Hassan, Mukherjee, Nagajyothi, Weiss and Petkova2006; Corral et al., Reference Corral, Guerrero, Cuervo, Girones and Fresno2013), and TXA2 is induced exacerbating cardiomyocyte apoptosis, facilitating cytokine biosynthesis by monocytes, and leading to endothelial and platelet activation, aggregation and degranulation (Corral et al., Reference Corral, Guerrero, Cuervo, Girones and Fresno2013).

Virulence factors of T. cruzi

T. cruzi possesses a wide variety of virulence factors that allow it to reproduce and effectively evade host defences and generate damage or morbidity in the affected individual (Brown et al., Reference Brown, Cornforth and Mideo2012), these virulence factors are described below.

Many studies have focused on finding and characterizing the molecules involved in the process of infection by T. cruzi, but little is known about the molecules involved in cellular invasion by extracellular amastigotes. Recently, a group of amastine proteins was reported, for which there are four subfamilies (alpha, beta, delta and gamma), of which the beta and delta proteins are found in T. cruzi (Jackson, Reference Jackson2010). These proteins are located on the amastigote cell surface and in strain G were demonstrated to increase the rate of differentiation towards metacyclic trypomastigotes. Studies with recombinant proteins both in vitro and in vivo demonstrated that these proteins also participated in the adhesion to and invasion of host cells by the amastigotes, and intracellular survival of the parasites by maintaining the pH inside parasitic vacuoles potentially via a mechanism that involves proton and ion trafficking through the membrane (Cruz et al., Reference Cruz, Souza-Melo, da Silva, DaRocha, Bahia, Araújo, Teixeira and Mortara2012).

One of the most important virulence factors in T. cruzi is the TS, which allow for sialylation of T. cruzi glycoconjugate membranes at terminal positions. There are two isoforms of TS identified in T. cruzi, one of which is enzymatically active (aTS) and the other is inactive (iTS), but still retains the ability to bind to sugar substrates. These proteins are secreted into the bloodstream, where they are systemically distributed and induce modifications in host cells by sialylation (Freire-de-Lima et al., Reference Freire-de-Lima, Fonseca, Oeltmann, Mendonça-Previato and Previato2015). After intracellular replication of the parasites and transformation into trypomastigotes, a large amount of TS is released into the cytoplasm of the parasitized cell, which is then lysed leading to an increase in the concentration of systemic TS that cannot be efficiently combated by antibodies. The amount of enzyme secreted correlates with the virulence of the strain and high levels of TS are associated with a wide variety of abnormalities during the early stages of infection, including thymocyte depletion (thymocyte apoptosis), the absence of germinal centres in secondary organs, thrombocytopenia and erythropenia (Freire-de-Lima et al., Reference Freire-de-Lima, Fonseca, Oeltmann, Mendonça-Previato and Previato2015; San Francisco et al., Reference San Francisco, Gutierrez, Neira, Munoz, Sagua, Araya, Andrade, Zailberger, Catalán, Remonsellez, Vega and González2017) (Fig. 3).

Fig. 3. Virulence factors and immunological response elicited by T. cruzi infection.

TS also inhibit lymphocyte proliferation via IL-2 signalling, and are associated with the ability of the parasite to reduce IL-2Ra expression and IL-2 production by T cells. In addition, they can manipulate the maturation of CD4+ cells inducing non-protective Th2 genotypes in naive T cells while downregulating Th1 cells by IL-10 induction/expression during antigenic presentation by host cell antigens (ACP) to favour the survival and infection of the parasite. Additionally, CD8+ T cells from infected animals have been found to be highly sialylated, which reduces their ability to infiltrate tissues. This provides a dual benefit to the parasite, since it favours its intracellular replication while preventing extensive tissue damage of the infected tissue (Tarleton, Reference Tarleton2015; San Francisco et al., Reference San Francisco, Gutierrez, Neira, Munoz, Sagua, Araya, Andrade, Zailberger, Catalán, Remonsellez, Vega and González2017).

Similarly, the parasite is covered by a group of mucin glycoconjugates, which are classified depending on whether they are present in the vector (TcMUC) or in the host (TcSMUG). These mucin glycoconjugates offer protection against the vector or host defences, thereby assuring cellular invasion (Osorio et al., Reference Osorio, Rios, Gutierrez and Gonzalez2012). Surface antigens also exist that are related to these glycoconjugates, such as the mucin-like gp35/50 surface antigen that compromises cellular invasion and the trypomastigote small surface antigen (TSSA) of the trypomastigote. Genetic analysis of the TSSA revealed a strong relationship with TcMUC and variations among the sequences of the six DTUs of T. cruzi (TcI–TcVI). However, unlike TcMUC, TSSA is a hypoglycosylated molecule that participates in the infectivity of the trypomastigote by acting as an adhesion molecule between the host and parasite and playing a role in amastigogenesis. There are two variants of TSSA, one of which shows adhesive properties (TSSA-CL), and the other is non-adhesive (TSSA-Sy). TSSA is a potential target as a diagnostic and therapeutic effectiveness agent (Yoshida, Reference Yoshida2006; Balouz et al., Reference Balouz, Melli, Volcovich, Moscatelli, Moroni, González, Ballering, Bisio, Ciocchini and Buscaglia2017; Cámara et al., Reference Cámara, Cánepa, Lantos, Balouz, Yu, Chen, Campetella, Mucci and Buscaglia2017).

The T. cruzi parasite has mechanisms for the evasion of reactive species of oxygen and hydrogen (e.g. O2 and H2O2), produced mainly by macrophages, that generate direct oxidative damage. These defence mechanisms are mainly coordinated by five different types of peroxidase (TcGPXI, TcGPXII, TcCPX, TcMPX and TcAPX), the first two conferring protection against exogenous hydroperoxides, and TcCPX, TcMPX, and small chains of organic hydroperoxides and TcAPX conferring protection against H2O2. T. cruzi also possesses an iron superoxide dismutase (Fe-SOD) that prevents damage by O2 from mitochondrial, cytosolic or glycosomal sources (Osorio et al., Reference Osorio, Rios, Gutierrez and Gonzalez2012; Malvezi et al., Reference Malvezi, Da Silva, De Freitas, Lovo-Martins, Tatakihara, Zanluqui, Neto, Goldenberg, Bordignon, Yamada-Ogatta, Martin-Pinge, Cecchini and Pinge-Filho2014).

T. cruzi possesses other molecules that contribute to the evasion of host immune responses, such as those that inhibit the coupling of complement molecules. Complement regulatory proteins (CRPs) expressed only in trypomastigotes, inhibit the classical and alternative pathways by binding to C3b and C4b. For example, trypomastigote decay accelerating factor (T-DAF) interferes with the coupling of C3 in both pathways, and its metacyclic trypomastigote form is able to induce blood cells to produce microvesicles that bind to the surface of the parasite and stabilize it to C3. Similarly, the complement C2 receptor inhibitor trispanning protein (CRIT) inhibits complement activation via lectin, while calreticulin (TcCRT) captures C1 molecules and uses them as host cell recognition molecules to the benefit of the parasite (Osorio et al., Reference Osorio, Rios, Gutierrez and Gonzalez2012; Henrique et al., Reference Henrique, Marques, da Silva, de Oliveira, Rodrigues, Gomez-Hernandez, Ramirez, Ferreira, Nogueira and Norris2016).

Other molecules such as calpain, a calcium-dependent lysosomal peptide cysteine, present in the endosome–lysosome system of the epimastigotes, on the epimastigote cell surface and in the amastigote–trypomastigote transitional forms, is secreted from the flagellar pocket and intervenes in the processes of cytoskeleton remodelling, proliferation, differentiation and regulation of cellular calcium. Calpain operates through the cleavage of the high molecular weight kininogen protein, which stimulates the release of calcium through inositol triphosphate, among other molecules (de Souza et al., Reference de Souza, de Carvalho and Barrias2010; Branquinha et al., Reference Branquinha, Marinho, Sangenito, Oliveira, Goncalves, Ennes-Vidal, d'Avila-Levy and Santos2013). Oligopeptidase B, a serine endopeptidase cytosolic, secreted by trypomastigotes, has also been implicated in the induction of calcium release during invasion of the parasite (Burleigh et al., Reference Burleigh, Caler, Webster and Andrews1997; Burleigh and Woolsey, Reference Burleigh and Woolsey2002; de Souza et al., Reference de Souza, de Carvalho and Barrias2010).

Finally, alteration in the metabolism of phospholipids has recently been reported as another possible mechanism by which parasites interact with the host. For example, molecules such as phospholipase A1 (PLA1) have been shown to participate in parasite–cell interactions prior to cell invasion through the generation of lipids that act as secondary messengers and co-activate kinase C (Belaunzaran et al., Reference Belaunzaran, Lammel, Gimenez, Bott, Durante de Isola, Wilkowsky and Barbieri2013). The expression of phosphatidylinositol phospholipase C (TcPI-PLC) correlates with the decrease in phosphatidylinositol-4,5-bisphosphate (PIP2) and the increase in its product inositol-1,4,5-triphosphate in the cell host demonstrating that overexpression of TcPI-PLC can inhibit the progression of trypomastigotes to amastigotes (Okura et al., Reference Okura, Fang, Salto, Singer, Docampo and Moreno2005; Osorio et al., Reference Osorio, Rios, Gutierrez and Gonzalez2012).

The main limitations in the identification of these virulence factors is that there is no clue if these are DTU-dependent. Most of the studies include merely the Y strain (typed as TcII). Therefore, future studies should establish if these virulence factors are deviated by the massive genetic diversity among T. cruzi taxon.

Host factors

Polymorphisms among certain host factors, such as those that affect the expression of cytokines involved in the immune response, can play an important role in the course of a disease by attenuating the defensive capacity of the host against an invading pathogen. The relationship between CCC and cytokine expression (e.g. for IL-1, IL-10, IL-12, IL-17 and IL-18) has been studied, along with preliminary studies to analyse the relationship between this disease and other polymorphisms among host immune factors (Leon et al., Reference Leon Rodrigez, Carmona, Echeverría, González and Martin2016).

For IL-1, a proinflammatory cytokine that has been implicated in the mediation of both acute and chronic manifestations of CCC disease, has been reported to offer protection in terms of the haplotypes IL-1A, IL-1B and IL-1RN (Flórez et al., Reference Flórez, Zafra, Morillo, Martín and González2006). For IL-10, polymorphisms that lead to reduced expression, such as IL-1082G/A, are associated with the development of CCC. For IL-17, a proinflammatory cytokine produced by CD4+ T cells, some studies have shown that lower expression of this cytokine correlates with cardiac manifestations, whereas associations between certain polymorphisms (rs2275913, rs763780) have been related to the severity of left ventricular systolic function in patients with CCC (Costa et al., Reference Costa, Moreira, Menezes, Silva, Dutra, Rocha and Gollob2009; Magalhães et al., Reference Magalhães, Villani, Nunes, Gollob, Rocha and Dutra2013; Reis et al., Reference Reis, Sakita, de Moraes, Aquino, Macedo, Mazini, Sell, de Oliveira, Bulgarelli and Laguila2017). As for IL-18, reduced expression of this cytokine has been associated with the early stage response to CCC (rs360719*C) by permitting the activation of transcription factor OCT-1; however, certain polymorphisms (rs5744258, rs360722) showed no statistically significant relationship with susceptibility to disease, and other studies found that rs2043055 was associated with the modulation of Chagas disease severity (Esper et al., Reference Esper, Utsch, Brant, Teixeira, Vieira, Machado, Soriani, Arantes, Campos, Pinho, Souza and Tanowitz2014; Nogueira et al., Reference Nogueira, Frade, Ianni, Laugier, Pissetti, Cabantous, Baron, Peixoto-Gde, Borges-Ade, Donadi, Marin-Neto, Schmidt, Dias, Saba, Wang, Fragata, Sampaio, Hirata, Buck, Mady, Martinelli, Lensi, Siqueira, Pereira, Rodrigues, Kalil, Chevillard and Cunha-Neto2015). Further studies are therefore required to investigate the relationships between host polymorphisms and CCC disease.

Similarly, polymorphisms among chemokines have been implicated in the pathology of CCC disease. Chemokines play an important role in the recruitment of inflammatory cells to heart tissue in studies of mice with both acute and chronic myocarditis. Variations in CCL2, a cytokine that participates in the elimination of parasites through NO-dependent pathways, have been associated with the development of CCC. Similarly, polymorphisms in CCR5, which is essential in the control of acute infections and in the maintenance of inflammation that causes tissue damage, have been related to CCC disease severity (CCR5 rs1799988) and other polymorphisms are more frequently associated with asymptomatic patients (CCR5 59029A/G). Furthermore, the pleomorphism rs10336 in CXCL9 [105–110] correlated with the diminution of the intensity of myocarditis. By contrast, variations in CCL4, CCL5, CCL17, CCL19 and CXCR3 showed no association with CCC (Fernández-Mestre et al., Reference Fernández-Mestre, Montagnani and Layrisse2004; Nogueira et al., Reference Nogueira, Santos, Ianni, Fiorelli, Mairena, Benvenuti, Frade, Donadi, Dias, Saba, Wang, Fragata, Sampaio, Hirata, Buck, Mady, Bocchi, Stolf, Kalil and Cunha-Neto2012).

Variations in HLA supertypes (such as HLA-A1, A2 and A24, which are present in CD8+ T cells where most of their epitopes are restricted to the HLA-A allele) have also been found to relate to disease pathology (Lasso et al., Reference Lasso, Beltrán, Guzmán, Rosas, Thomas, López, González, Cuéllar and Puerta2016), with polymorphisms in HLA I and HLA II being reported to be associated with CCC (Deghaide et al., Reference Deghaide, Dantas and Donadi1998). Of the HLA I genes, it was shown that variations in HLAC*03 have a strong association with CCC compared with asymptomatic patients. Similarly, DPB1*0401 presents a higher frequency in affected patients compared with asymptomatic patients, with asymptomatic patients showing a higher frequency of DPB1*0101 (Colorado et al., Reference Colorado, Acquatella, Catalioti, Fernandez and Layrisse2000; Layrisse et al., Reference Layrisse, Fernandez, Montagnani, Matos, Balbas, Herrera, Colorado, Catalioti and Acquatella2000). As for the HLA II genes, a comparison was made between alleles DRB1 and DQB1 and polymorphisms DRB1*14 and DQB1*0303 were found to be protective against the chronic phase of the disease. In addition, the DRB1*01, DRB1*08 and DQB1*0501 polymorphisms showed a higher frequency in patients with arrhythmia and congestive heart failure, whereas the DRB1*1501 polymorphism presented a lower frequency in such patients. These findings suggested that the HLA II genes may be associated with the development of chronic infection and cardiac tissue damage. Similarly, polymorphisms in HLA-DQ1 are considered to confer susceptibility, whereas HLA-DQ7 confers protection against the development of CCC disease (Deghaide et al., Reference Deghaide, Dantas and Donadi1998; Cunha-Neto and Chevillard, Reference Cunha-Neto and Chevillard2014).

Finally, for TNF, the TNFA-308 polymorphism and variations in the microsatellites of TNF-α from patients with end-stage CCC correlated with significantly shorter survival times compared with individuals who did not possess these variations (Drigo et al., Reference Drigo, Cunha-Neto, Ianni, Faé, Nunes, Buck, Mady, Kalil, Goldberg, Cardoso and Braga2006). Furthermore, variants in the promoter region of the IKBL gene (IKBL-62A/T, IKBL-262A/G) were associated with susceptibility to CCC (Ramasawmy et al., Reference Ramasawmy, Faé, Cunha-Neto, Borba, Ianni, Mady, Goldberg and Kalil2008). It has been recently suggested that the parasympathetic nervous system and the endocrine system may play important roles in the clinical manifestations associated with CCC (Roggero et al., Reference Roggero, Rosa Perez, Pollachini, Raquel Villar, Wildmann, Besedovsky and del Rey2016; Savino, Reference Savino2017). Further investigations are now needed on large study populations to determine the frequency of certain polymorphisms among individuals and the roles of host factor gene variations on the susceptibility and severity of CCC disease pathology (Prata, Reference Prata2001).

Cytokine profile during T. cruzi infection

The expression of cytokines and their kinetics are key factors in the development and progression of disease. At present, the pathophysiological mechanisms of Chagas’ disease have not been completely elucidated; however, two hypotheses have been established. The first relates to the early stages of infection where an appropriate inflammatory response is beneficial but a dysregulated response allows for tissue damage. In the acute phase of infection, it has been proposed that the immune response of the host may be associated with aggressive autoimmunity. The second hypothesis is based on an autoreactive process in response to the lack of adequate immune modulation between excessive proinflammatory cytokines and the loss of anti-inflammatory cytokines that plays an important role in the progression of human Chagas disease from asymptomatic to severe forms (cardiomyopathy and megavisceral syndromes) and is associated with molecular mimicry (Nagib et al., Reference Nagib, Dutra, Chiari, Conceição and Machado2007; Pissetti et al., Reference Pissetti, Correia, de Oliveira, Llaguno, Balarin, Silva-Grecco and Rodrigues2011; Longhi et al., Reference Longhi, Tasso, Gomez, Atienza, Bonato, Chiale, Perez, Buying, Pinilla, Judkowski, Balouz, Buscaglia and Santos2014).

During the acute phase of the disease, it has been suggested that the immune response employs three mechanisms to counteract the infection. The first is the detection and destruction of the parasite via macrophages and dendritic cells. The second involves the activation of dendritic cells and macrophages that initiate the presentation and activation of specific antigens to elicit immune responses. The third mechanism is the detection of infection by non-hematopoietic cells, which play a major role in protecting against the invasion of T. cruzi (Poveda et al., Reference Poveda, Fresno, Gironès, Martins-Filho, Ramírez, Santi-Rocca, Marín-Neto, Morillo, Rosas and Guhl2014).

In the chronic phase, antigens that are produced by the autoimmune response are mediated by T cells, as an important step in the progression of the disease. Several studies in children have found T cells to be predominant proinflammatory and cellular monocyte regulators. CD8+ T cells play an important role in combatting intracellular pathogens. This is due to the fact that this cell type is able to recognize infected cells, when such a function is eliminated or inhibited, there is no parasitic control in the early phase of the disease leading to the exacerbation of infection and the onset of chronic disease (Sanmarco et al., Reference Sanmarco, Visconti, Eberhardt, Ramello, Ponce, Spitale, Vozza, Bernardi, Gea, Minguez and Aoki2016).

Immune mechanisms that operate to control the parasite prior to intracellular infection are controlled by the CD4+ and CD8 cellular responses, which inhibit the replication of T. cruzi as demonstrated in vitro. These cells and the protective functions of IL-2, IFN-γ and TNF-α produced by Th1 cells, have been shown to be associated with heart disease. IL-5, IL-10 and IL-13 regulate the inflammatory humoral response and the stimulation of IgE, eosinophils and mastocytes (Teixeira et al., Reference Teixeira, Hecht, Guimaro, Sousa and Nitz2011).

A previous study detected a switch between the anti-inflammatory cytokines IL-13, IL-5 and IL-10 and proinflammatory cytokines IL-2, IL-6, IL-9 and IL-12 in 109 seropositive patients and 21 seronegative controls, who were classified into two groups, CARD (heart disease) and NON-CARD (no heart disease), and a clear lack of immune modulation was reported. By measuring the mean fluorescence intensity of IL-12, IFN-γ, IL-1, IL-6 and IL-9 for discriminant analysis of principal components (DAPC), a cluster was observed for the CARD patients that was not present with the NON-CARD patients, potentially indicating that some NON-CARD patients may be predisposed to developing cardiomyopathy or mega-viscera syndrome explaining why the cytokine levels are not homogeneous for this group and suggesting that these cytokines could be used in combination as progression markers (Poveda et al., Reference Poveda, Fresno, Gironès, Martins-Filho, Ramírez, Santi-Rocca, Marín-Neto, Morillo, Rosas and Guhl2014).

Role of cytokines in the genetic variability of T. cruzi DTUs

Previous studies to analyse the association between T. cruzi genetic variability and the different clinical manifestations of Chagas disease found an association between TcI and cardiomyopathy, and between TcII, TcV and TcVI and mega-visceral syndrome. We used sera from CCC patients infected with different DTUs (20 TcI, 20 TcII and 15 mixed TcI + TcII) to observe whether the genetic variability of the parasite was associated with the pathogenesis of Chagas disease. A proinflammatory profile was observed for all groups as expected, and this profile was similar to that identified for the cardiac group. However, higher levels of cytokines were found in the TcII and mixed groups compared with the TcI group, where the levels did not exceed 50%. By contrast, we found higher levels of the cytokines IL-6 for TcI, IL-1 for TcII and IL-22 for the mixed TcI/TcII group (Rassi et al., Reference Rassi, Rassi and Marin-Neto2009). This is interesting since it is known that IL-6 is secreted by T cells and macrophages, IL-1 by macrophages and lymphocytes, and IL-22 by dendritic cells and T cells (Rojas. et al., Reference Rojas, Anaya, Gómez, Aristizabal, Cano and Lopera2017) (Table 1).

Table 1. Association between the T. cruzi DTUs and the related clinical form and immune response

A recent study showed that the immune profile relating to the cardiac inflammatory response involved the expression of TNF, IL-2, IL-10 and IFN-γ in human cardiac cell infiltrate samples, which may suggest that these factors play an important role in the variable susceptibility to the chronic phase of the disease. Other studies demonstrated the presence of IL-2, IL-4 and IL-6 in infected cardiac tissue. Even in patients with ventricular dysfunction, IL-10, IFN-γ, IL-6, TNF and IL-1 have been reported to increase plasma levels. This suggests that there is a relationship between the secretory response of T cells in the immunological profile of infected heart tissue (Vicco et al., Reference Vicco, Ferini, Rodeles, Cardona, Bontempi, Lioi, Beloscar, Nara, Marcipar and Bottasso2013; Rodríguez et al., Reference Rodríguez, Marigorta and Navarro2014). However, a study by Vicco and colleagues in Wistar mice showed that the administration of diluted phosphorus allowed for some modulation of inflammation in the cardiac tissue via IFN-γ and TNF-α (Ferreira et al., Reference Ferreira, Portocarrero, Brustolin, Ciupa, Massini, Aleixo and de Araújo2017).

In contrast to previous studies that showed that patients with less aggressive forms of cardiomyopathy produced higher levels of IL-17 (Guedes et al., Reference Guedes, Gutierrez, Silva, Dellalibera-Joviliano, Rodrigues, Bendhack, Rassi, Schmidt, Maciel, Marin- Neto and Silva2012), we hypothesize that DTU-specific recognition by the immune system (antibodies, B cells or T cells), could lead to the differential responses observed. Our results suggest that patients with more severe cardiomyopathy would be those with TcI, followed by those with a mixed infection, and finally those infected with TcII. This is in accordance with a descriptive analysis performed by our group where we detected that patients infected with the TcI DTU displayed more cardiac alterations than those infected with TcII (Ramírez et al., Reference Ramírez, Guhl, Rendón, Rosas, Marin-Neto and Morillo2010).

Finally using IFN-γ, IL-12, IL-22 and IL-10 for DAPC analysis, we found clusters for patients infected with TcI and mixed TcI + TcII, suggesting that there is a likely association between the genetic variability of T. cruzi (TcI, TcII and mixed TcI/TcII) and the levels of some cytokines. The sympathetic nervous system is thought to play an important role in the survival of T. cruzi infection based on studies in mice (C57B1/6) given the possible participation of mechanisms that upregulate the production of proinflammatory cytokines, as well as cellular immune responses and the restriction of parasitic proliferation (Roggero et al., Reference Roggero, Rosa Perez, Pollachini, Raquel Villar, Wildmann, Besedovsky and del Rey2016).

T. cruzi heterogeneity and CCC

The classification of T. cruzi parasites is important in defining the biological, clinical and pathological characteristics associated with specific populations of T. cruzi (Mantilla et al., Reference Mantilla, Zafra, Macedo and González2010; Dias et al., Reference Dias, Ramos, Gontijo, Luquetti, Shikanai-Yasuda, Coura, Torres, Melo, Almeida, Oliveira, Silveira, Rezende, Pinto, Ferreira, Rassi, Fragata, Sousa, Correia, Jansen, Andrade, Britto, Pinto, Rassi, Campos, Abad-Franch, Santos, Chiari, Hasslocher-Moreno, Moreira, Marques, Silva, Marin-Neto, Galvão, Xavier, Valente, Carvalho, Cardoso, Silva, Costa, Vivaldini, Oliveira, Valente, Lima and Alves2016). Despite intensive research into the molecular epidemiology of T. cruzi, few studies have investigated the association between the genetic heterogeneity of T. cruzi and the clinical outcomes of Chagas disease. The DTUs of T. cruzi involved in an infection can alter the humoral response affecting the pathogenesis of the disease (Santi-Rocca et al., Reference Santi-Rocca, Fernandez-Cortes, Chillon-Marinas, Gonzalez-Rubio, Girones, Fresno and Martin2017). For this reason, different authors have proposed the use of lineage-specific serology markers to detect the serological profile of an infection according to each DTU. These assays have clearly demonstrated that the immune response elicited by each DTU may be different and highlights the need for further research in this field (Zingales et al., Reference Zingales, Andrade, Briones, Campbell, Chiari, Fernandes, Guhl, Lages-Silva, Macedo, Machado, Miles, Romanha, Sturm, Tibayrenc and Schijman2009). TSSA antigen and B-cell epitopes have been investigated for this purpose of developing specific serology markers but experiments were hindered by the lack of specificity of the in vitro assays (Bhattacharyya et al., Reference Bhattacharyya, Brooks, Yeo, Lewis, Llewellyn, Miles and Carrasco2010, Reference Bhattacharyya, Falconar, Luquetti, Costales, Grijalva, Lewis, Messenger, Tran, Ramirez, Guhl, Carrasco, Diosque, Garcia, Litvinov and Miles2014) (Fig. 4).

Fig. 4. Overview of the geographical distribution of T. cruzi DTUs associated with CCC across the American continent.

Several studies have provided evidence of the existence of a relationship between T. cruzi DTUs and clinical manifestations. TcI has been reported to be the most abundant DTU across the American continent, being detected among a wide range of reservoirs and triatomines, in which its presence is derived predominantly from sylvatic rather than domestic transmission cycles. The infection of humans by this DTU is concentrated in the north of South America reaching the centre of America, and is mainly associated with chagasic cardiomyopathy, predominantly in the domestic cycle (Zingales et al., Reference Zingales, Andrade, Briones, Campbell, Chiari, Fernandes, Guhl, Lages-Silva, Macedo, Machado, Miles, Romanha, Sturm, Tibayrenc and Schijman2009; Leiby et al., Reference Leiby, Nguyen, Proctor, Townsend and Stramer2017). It has been reported that this particular DTU plays an important role in severe forms of chagasic heart disease (Guhl, Reference Guhl2013; Guhl and Ramírez, Reference Guhl and Ramírez2013). In a study carried out in Argentina of 239 patients with a diagnosis of severe myocarditis, TcI was detected in 4.2% of the patients from either blood samples, biopsies or the organs from those who underwent a transplant (Burgos et al., Reference Burgos, Diez, Vigliano, Bisio, Risso, Duffy, Cura, Brusses, Favaloro, Leguizamon, Lucero, Laguens, Levin, Favaloro and Schijman2010; Guhl, Reference Guhl2013). In Colombia, similar studies showed that TcI caused more cardiac alterations that TcII across a large cohort of chronic symptomatic patients (Ramírez et al., Reference Ramírez, Guhl, Rendón, Rosas, Marin-Neto and Morillo2010).

Studies on TcII have shown that this DTU of T. cruzi prevails in the central and southern regions of America, and is related to transmission of the domestic cycle. TcII has been associated with the clinical manifestations of moderate chagasic cardiomyopathy, concomitant with mega syndromes such as megacolon and megaesophagus, also associated with blood donors (Zingales et al., Reference Zingales, Andrade, Briones, Campbell, Chiari, Fernandes, Guhl, Lages-Silva, Macedo, Machado, Miles, Romanha, Sturm, Tibayrenc and Schijman2009; Guhl and Ramírez, Reference Guhl and Ramírez2013; Leiby et al., Reference Leiby, Nguyen, Proctor, Townsend and Stramer2017). In addition, Bisio and coworkers confirmed that TcII was present in two patients with cardiac manifestations. However, these estimations are not absolute and patients across the continent with CCC can be infected by TcII (Burgos et al., Reference Burgos, Bisio, Duffy, Levin, Schijman, Altcheh, Burgos Freilij, Valadares, Freitas, Macedo, Seidenstein, Macchi and Piccinali2007).

In endemic areas of the Amazon region of Brazil, and in eastern Colombia and Venezuela, TcIV is considered to be the predominant DTU responsible for most of the acute diseases caused by this DTU (Carrasco et al., Reference Carrasco, Segovia, Llewellyn, Morocoima, Urdaneta-Morales, Martínez, Garcia, Rodríguez, Espinosa, de Noya, Díaz-Bello, Herrera, Fitzpatrick, Yeo, Miles and Feliciangeli2012; Guhl and Ramírez, Reference Guhl and Ramírez2013; Segovia et al., Reference Segovia, Carrasco, Martínez, Messenger, Nessi, Londoño, Espinosa, Martínez, Mijares, Bonfante-Cabarcas, Lewis, de Noya, Miles and Llewellyn2013; Monteiro et al., Reference Monteiro, Peretolchina, Lazoski, Harris, Dotson, Abad-Franch, Tamayo, Pennington, Monroy, Cordon-Rosales, Salazar-Schettino, Gómez-Palacio, Grijalva, Beard and Marcet2013a; Reference Monteiro, Margioto-Teston, Gruendling, dos Reis, Gomes, de Araújo, Bahia, Magalhães, de Oliveira-Guerra, Silveira, Toledo and Vale-Barbosa2013b; Margioto Teston et al., Reference Margioto Teston, de Abreu, Abegg, Gomes and de Ornelas Toledo2017). TcIV has also been strongly incriminated with lethal cases relating to oral transmission in Colombia and Brazil (Monteiro et al., Reference Monteiro, Peretolchina, Lazoski, Harris, Dotson, Abad-Franch, Tamayo, Pennington, Monroy, Cordon-Rosales, Salazar-Schettino, Gómez-Palacio, Grijalva, Beard and Marcet2013a; Reference Monteiro, Margioto-Teston, Gruendling, dos Reis, Gomes, de Araújo, Bahia, Magalhães, de Oliveira-Guerra, Silveira, Toledo and Vale-Barbosa2013b; Ramírez et al., Reference Ramírez, Montilla, Cucunubá, Floréz, Zambrano and Guhl2013; Dario et al., Reference Dario, Rodrigues, Barros, Xavier, Roque, Jansen and D'Andrea2016; Hernández et al., Reference Hernández, Cucunubá, Flórez, Olivera, Valencia, Zambrano, León and Ramírez2016). As for TcV and TcVI, comparative genetic studies proposed that these DTUs are TcII and TcIII hybrids that correlate with chagasic cardiomyopathy and megavisceral syndrome in the southern cone of the American continent (Zingales et al., Reference Zingales, Andrade, Briones, Campbell, Chiari, Fernandes, Guhl, Lages-Silva, Macedo, Machado, Miles, Romanha, Sturm, Tibayrenc and Schijman2009; Guhl and Ramírez, Reference Guhl and Ramírez2013). In a study from Argentina, TcV or TcII/V/VI were found to be the most prevalent DTUs among 226 of 239 patients studied (89.9%). Whereas, TcV was present in 90.9% of the TcII/V/VI group samples studied that correlated with Chagas’ moderate chronic heart disease in the study by Burgos and colleagues (Burgos et al., Reference Burgos, Diez, Vigliano, Bisio, Risso, Duffy, Cura, Brusses, Favaloro, Leguizamon, Lucero, Laguens, Levin, Favaloro and Schijman2010).

Another study revealed that in the southern region of Latin America (in particular Argentina), blood samples and cultures isolated from patients with CCC were predominately of the TcII, TcV and TcVI T. cruzi DTUs. To date, in this geographic location, TcI has not been considered the dominant DTU in heart disease (Cura et al., Reference Cura, Bisio, Duffy, Schijman, Lucero, Formichelli, Brusés, Merino, Oshiro, Sosa-Estani, Burgos, Lejona, Anchart, Hernández, Severini, Velazquez, Lattes, Altcheh, Freilij, Diez, Nagel, Vigliano, Favaloro and Favaloro2012). However, in terms of clinical associations, descriptive molecular epidemiology studies linked TcI with severe forms of myocarditis in cardiac samples from CCC patients in Argentina and no specific clinical manifestations related to T. cruzi DTUs in Bolivian CCC patients showing the pleomorphism of T. cruzi (Moncayo and Yanine, Reference Moncayo and Yanine2006; Ramírez et al., Reference Ramírez, Guhl, Umezawa, Morillo, Rosas, Marin-Neto and Restrepo2009; Burgos et al., Reference Burgos, Diez, Vigliano, Bisio, Risso, Duffy, Cura, Brusses, Favaloro, Leguizamon, Lucero, Laguens, Levin, Favaloro and Schijman2010; del Puerto et al., Reference del Puerto, Nishizawa, Kikuchi, Iihoshi, Roca, Avilas, Gianella, Lora, Velarde, Gutierrez, Renjel, Miura, Higo, Komiya, Maemura and Hirayama2010). The direct detection of T. cruzi DTUs in the blood of CCC patients was established by amplification of the 24Sα rDNA divergent domain and the mitochondrial housekeeping genes (Mantilla et al., Reference Mantilla, Zafra, Macedo and González2010). In this study, molecular characterization of T. cruzi DTUs showed that most of the patients were infected with TcI and some were infected with TcII (9.9%). Recently, a new approach for T. cruzi DTU detection in CCC patients has been developed that showed that TcI was the predominant DTU and TcII was also detected, furthermore, the genetic characteristics of the TcII parasites found in Colombia were similar to those of the TcII parasites found in Bolivia and Chile (Mantilla et al., Reference Mantilla, Zafra, Macedo and González2010). Regarding the genetic variability of the parasite, prognosis markers based on mitochondrial genes are being developed, since specific mutations in these genes can trigger complications in the chronic phase of the disease in asymptomatic patients (Carranza et al., Reference Carranza, Valadares, D’Ávila, Baptista, Moreno, Galvão, Chiari, Sturm, Gontijo, Macedo and Zingales2009).

Despite the genetic variability, it is important to consider the presence of T. cruzi clones that have been detected in different tissues. Several studies have demonstrated specific histotropism of T. cruzi in mice showing differences in the pathological, immunological and clinical features that the parasite can elicit in the host (Carareto et al., Reference Carareto, Manoel-Caetano, Aparecida, Silva, Borim and Miyazaki2008; Ramírez et al., Reference Ramírez, Guhl, Rendón, Rosas, Marin-Neto and Morillo2010; Cruz et al., Reference Cruz, Santos-Miranda, Monti-Rocha, Machado, Sales, Campos and Roman-Campos2016; Leon et al., Reference Leon, Ramirez, Montilla, Vanegas, Castillo and Parra2017). Moreover, some authors have shown that the T. cruzi population in a patient's bloodstream may differ from the parasite population that causes tissue damage (Vago et al., Reference Vago, Andrade, Leite, d'Avila-Reis, Macedo, Adad, Tostes, Moreira, Filho and Pena2000; Macedo et al., Reference Macedo, Machado, Oliveira and Pena2004). Differences were found in T. cruzi populations in the bloodstreams of patients with CCC and of chagasic patients without cardiomyopathy (Venegas et al., Reference Venegas, Coñoepan, Pichuantes, Miranda, Apt, Arribada, Zulantay, Coronado, Rodriguez, Reyes, Solari and Sanchez2009). Microsatellite analyses have shown multiclonality in samples from the heart and bloodstream of infected patients demonstrating that specific populations of T. cruzi may determine the disease outcome (Burgos et al., Reference Burgos, Bisio, Duffy, Levin, Schijman, Altcheh, Burgos Freilij, Valadares, Freitas, Macedo, Seidenstein, Macchi and Piccinali2007; Valadares et al., Reference Valadares, Pimenta, de Freitas, Duffy, Bartholomeu, Oliveira, Chiari, Moreira, Filho, Schijman, Franco, Machado, Pena and Macedo2008). Finally, studies from clones obtained from hemoculture from one patient (chronic symptoms) showed divergent multilocus genotypes, reinforcing this hypothesis (Ramírez et al., Reference Ramírez, Guhl, Messenger, Lewis, Montilla, Cucunuba and Llewellyn2012, Reference Ramírez, Montilla, Cucunubá, Floréz, Zambrano and Guhl2013). One group proposed a model of clonal histiotropism to explain how a composite of clones may be related to disease outcomes (Macedo and Segatto, Reference Macedo and Segatto2010). Recent reports from Colombia corroborate this premise since they describe cardiac biopsies infected with sylvatic genotypes and TcI circulating in the bloodstream of patients (Ramírez et al., Reference Ramírez, Guhl, Rendón, Rosas, Marin-Neto and Morillo2010; Zafra et al., Reference Zafra, Mantilla, Jacome, Macedo and Gonzalez2011). The case of mixed infections appears to be the rule rather than the exception in chagasic patients. It has been proposed that such mixed infections play a major role in Chagas pathogenicity, with some paradox traits: in animal models, infection by a ‘slow’ genotype and by a ‘fast’ genotype is faster than the faster genotype alone. This has been recently corroborated by Llewellyn et al. in Reference Llewellyn, Messenger, Luquetti, Garcia, Torrico, Tavares, Cheaib, Derome, Delepine, Baulard, Deleuze, Sauer, Miles and Michael2015 where they explored T. cruzi infection multiclonality in the context of age, sex and clinical profile among a cohort of chronic patients, as well as paired congenital cases from Cochabamba, Bolivia and Goias, Brazil using amplicon deep sequencing technology (Llewellyn et al., Reference Llewellyn, Messenger, Luquetti, Garcia, Torrico, Tavares, Cheaib, Derome, Delepine, Baulard, Deleuze, Sauer, Miles and Michael2015). Their conclusions showed that no specific association was found between the number and diversity of parasite genotypes in each patient with their age, sex or disease status. Also, they were able to detect the transmission of multiple parasite genotypes between mother and foetus. This clearly opens the window about the need to explore in deep the case of mixed infections across a well-characterized cohort of chagasic patients and demonstrate the pivotal role of multiclonality in a plausible severity of disease outcome (Llewellyn et al., Reference Llewellyn, Messenger, Luquetti, Garcia, Torrico, Tavares, Cheaib, Derome, Delepine, Baulard, Deleuze, Sauer, Miles and Michael2015).

Molecular epidemiology studies of T. cruzi have attempted to establish the effects of different DTUs in the clinical progression of Chagas disease. Several studies have shown the effect of genetic variability on the host immune response (Ramírez et al., Reference Ramírez, Guhl, Umezawa, Morillo, Rosas, Marin-Neto and Restrepo2009). However, a study carried out in the Chilean population, in which the DTUs TcI, TcII, TcV and TcVI were present and TcV predominated, sought to establish a relationship between the parasitic burden, the DTUs and the clinical manifestations of the disease, but found no correlation between the DTUs and the cardiac manifestations of the disease (Apt et al., Reference Apt, Zulantay, Saavedra, Araya, Arriagada, Arribada, Solari, Ortiz and Rodríguez2015). It had previously been established that the cardiopathologies in southern cone countries were caused by TcII, TcV and TcVI, but it has recently been demonstrated that TcI can also play an important role specifically in the severe cardiopathologies related to Chagas disease. Studies of cardiac biopsies from Argentinean patients revealed that patients with severe myocarditis were infected with TcI, whereas those with moderate or absent myocarditis were infected with TcII, TcV or TcVI (Burgos et al., Reference Burgos, Diez, Vigliano, Bisio, Risso, Duffy, Cura, Brusses, Favaloro, Leguizamon, Lucero, Laguens, Levin, Favaloro and Schijman2010). Furthermore, among patients with CCC, the TcIDOM genotype was most commonly found in the bloodstream, whereas sylvatic-like TcI parasites were most commonly found in cardiac biopsies. These results were consistent with reports from patients in Colombia, where the least and most prevalent TcI genotypes in adult patients with CCC were sylvatic-like TcI parasites and TcIDOM, respectively (Ramírez et al., Reference Ramírez, Guhl, Rendón, Rosas, Marin-Neto and Morillo2010; Hernández et al., Reference Hernández, Cucunubá, Flórez, Olivera, Valencia, Zambrano, León and Ramírez2016). These results suggest potential histotropism by TcI genotypes and the epidemiological importance of this DTU in the southern American countries, where cardiopathologies were previously thought to be caused primarily by TcII, TcV and TcVI.

The main problem in establishing the real picture of T. cruzi heterogeneity in Chagas disease patients is the low parasitic load in the chronic phase of the disease. Furthermore, a temporal variation pattern has been detected whereby the T. cruzi population may change at 10-day intervals (Sánchez and Ramírez, Reference Sánchez and Ramírez2013). Therefore, it is imperative to improve the current methodologies for strain typing. Recently, with the rise of next generation sequencing technologies, researchers have been able to deploy multilocus sequence typing (MLST) schemes to infer the genetic divergence of this parasite (Lauthier et al., Reference Lauthier, Tomasini, Rumi, D'Amato, Ragone, Diosque, Barnabé, Tibayrenc, Yeo, Lewis, Llewellyn, Miles and Basombrío2012; Messenger et al., Reference Messenger, Llewellyn, Bhattacharyya, Franzén, Lewis, Ramírez, Carrasco, Andersson and Miles2012). These methodologies have been applied to specific clinical phenotypes such as oral Chagas disease, which is an eminent public-health problem in those areas were vectorial transmission has been interrupted. In Colombia, six outbreaks of oral Chagas disease have been reported and strains isolated from these outbreaks were analysed by MLST schemes. The results showed a predominance of TcI in the cases with a foreseen infection of TcIV, suggesting the unlikely relatedness of sylvatic strains with the oral outbreaks (Hernández et al., Reference Hernández, Cucunubá, Flórez, Olivera, Valencia, Zambrano, León and Ramírez2016).

T. cruzi I has shown a relevant genetic heterogeneity and some authors have subdivided it into at least two near-clades (domestic and sylvatic TcI) (Ramírez and Hernández, Reference Ramírez and Hernández2017). However, other studies showed that only domestic TcI is a robust genotype across the American continent which clearly reflects that TcI isolates are highly variable (Zumaya-Estrada et al., Reference Zumaya-Estrada, Messenger, Lopez-Ordonez, Lewis, Flores-Lopez, Martínez-Ibarra, Pennington, Cordon-Rosales, Carrasco, Segovia, Miles and Llewellyn2012). Recent genomic approaches by whole genome sequencing and multi-SNP typing will most probably uncover lesser genetic subdivisions within it. This intra-DTU diversity is remarkable in the light of plausible histiotropism where has been revealed that domestic TcI circulates in the bloodstream and sylvatic TcI shows tropism for heart tissue (Burgos et al., Reference Burgos, Diez, Vigliano, Bisio, Risso, Duffy, Cura, Brusses, Favaloro, Leguizamon, Lucero, Laguens, Levin, Favaloro and Schijman2010). Such micro-molecular epidemiology tends to become routine in bacteria and has started in parasites (Wong et al., Reference Wong, Baker, Pickard, Parkhill, Page, Feasey, Kingsley, Thomson, Keane, Weill, Edwards, Hawkey, Harris, Mather, Cain, Hadfield, Hart, Thieu, Klemm, Glinos, Breiman, Watson, Kariuki, Gordon, Heyderman, Okoro, Jacobs, Lunguya, Edmunds, Msefula, Chabalgoity, Kama, Jenkins, Dutta, Marks, Campos, Thompson, Obaro, MacLennan, Dolecek, Keddy, Smith, Parry, Karkey, Mulholland, Campbell, Dongol, Basnyat, Dufour, Bandaranayake, Naseri, Singh, Hatta, Newton, Onsare, Isaia, Dance, Davong, Thwaites, Wijedoru, Crump, De Pinna, Nair, Nilles, Thanh, Turner, Soeng, Valcanis, Powling, Dimovski, Hogg, Farrar, Holt and Dougan2015). Such studies show that upper evolutionary units such as T. cruzi I are to broad units of analysis for refined epidemiological studies. Future studies must consider intra-DTU genetic variation and plausible disease outcome.

The analysis of specific haplotypes incriminated the TcI sylvatic-like strains in the oral cases of Chagas disease highlighting the need to improve current epidemiological surveillance systems in endemic areas to detect the invasion of sylvatic-like genotypes in domestic cycles of transmission (Ramírez et al., Reference Ramírez, Montilla, Cucunubá, Floréz, Zambrano and Guhl2013). This type of analysis was also conducted in the largest urban oral outbreak of Chagas disease ever reported in Caracas, Venezuela, where the authors were able to track the source of infection attributing it to TcI sylvatic-like strains (Segovia et al., Reference Segovia, Carrasco, Martínez, Messenger, Nessi, Londoño, Espinosa, Martínez, Mijares, Bonfante-Cabarcas, Lewis, de Noya, Miles and Llewellyn2013). This scenario proposes the relevance of the typing schemes to track the phylodynamics of T. cruzi and also indicates that some TcI DTUs are likely more susceptible to causing oral infections. However, some authors have proposed that TcI and TcII express different glycoproteins that facilitate their survival in the gastric mucosa leading to infection via the oral route (Yoshida et al., Reference Yoshida, Tyler and Llewellyn2011; Sánchez and Ramírez, Reference Sánchez and Ramírez2013). This area therefore requires further study.

Studies that encompass molecular epidemiology and molecular biology techniques, such as PCR, allow us to establish the relationship between different DTUs of T. cruzi and the clinical manifestations of the disease (including Chagas’ cardiomyopathy) in various populations. The findings of such studies indicate that certain genotypes of this parasite exert damage at the cellular level and therefore are not the same as those detected in the blood that cause parasitaemia and infection during the acute phase of the disease. In addition, the various serotypes of trypanosomes that cause non-heart-related pathologies and those that cause Chagas cardiomyopathy may differ, meaning that organisms belonging to a specific genotype may predominate in particular clinical manifestations. This suggests that specific genotypes may determine the clinical course of the disease (Guhl and Ramírez, Reference Guhl and Ramírez2013).

One interesting and important study was conducted by Santi-Rocca et al. in 2017 using syngeneic mice infected acutely or chronically with six DTUs, 66 parameters were analysed, including parasite tropism, organ and immune responses (local and systemic) and clinical presentations of CCC. The authors of that study found that the parasite genetic background consistently impacts most of these parameters, but they remain highly variable impeding reliable one-dimensional association with phases, strains and damage, but the use of multi-dimensional statistics overcame this extreme intra-group variability and revealed some pathophysiological patterns that accurately allow defining (i) the infection phase, (ii) the infecting parasite strains and (iii) organ damage type and intensity (Santi-Rocca et al., Reference Santi-Rocca, Fernandez-Cortes, Chillon-Marinas, Gonzalez-Rubio, Girones, Fresno and Martin2017). This study was very important towards the understanding of the association between T. cruzi genetic diversity, host genetics and disease outcome. However, the results are not merely conclusive and research must focus on this topic.

However, the existence of studies showing the non-existence of a relationship between the clinical manifestations of Chagas disease and the genetic heterogeneity of the parasite must be considered. According to one study, no lineage was found to have a significant association with any clinical manifestation in particular, nor exclusively with patients who presented with the chronic phase of the disease (del Puerto et al., Reference del Puerto, Nishizawa, Kikuchi, Iihoshi, Roca, Avilas, Gianella, Lora, Velarde, Gutierrez, Renjel, Miura, Higo, Komiya, Maemura and Hirayama2010).

Conclusions

This paper enhances our understanding of CCC as a multifactorial disease and highlights the heterogeneity of T. cruzi and how this heterogeneity relates to the pathology of disease. By collating the data from epidemiological studies, it becomes possible to gain an overview of which DTUs are predominant in particular geographic locations and which are associated with particular pathologies. These findings are useful in many respects including the development of control strategies to prevent the spread of the disease, for example, by preventing the migration of infected patients to non-endemic regions.

In addition, the clinical course of CCC disease depends on the immunological status of the host, as all aspects of innate immunity, as well as cytokine induction, are required to respond to T. cruzi infection. The pathology of disease is also affected by the response of the host to the virulence factors of the parasite. Processes such as pathogen recognition and parasite internalization into host cells all play a part in determining the pathophysiology of the disease. Despite intensive efforts of the scientific community, the studies to date are not conclusive regarding the true drivers of CCC. It is a complex interaction between the parasite and the host and for us is very speculative to propose responses based on the current data. We believe that the advent of new technologies such as QTL analysis using human and parasite genomes and also the cytokinome efforts would contribute to the future understanding of the CCC drivers. The new PacBio T. cruzi genome assembly and the high level of human genome annotation would be pivotal to solve this enigmatic question.

Acknowledgements

The authors thank Kate Fox, DPhil, from Edanz Group (www.edanzediting.com/ac) for editing a draft of this manuscript.

Financial support

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

Conflicts of interest

None.

Ethical standards

Not applicable.

Footnotes

*

Both authors contributed equally.

References

Andersson, B (2011) The Trypanosoma cruzi genome; conserved core genes and extremely variable surface molecule families. Research in Microbiology 162, 619625.Google Scholar
Angheben, A, Buonfrate, D, Gobbi, F, Bisoffi, Z, Boix, L, Pupella, S and Gandini, G (2015) Chagas disease and transfusion medicine: a perspective from non-endemic countries. Blood Transfusion 13, 540550.Google Scholar
Apt, W, Zulantay, I, Saavedra, M, Araya, E, Arriagada, K, Arribada, A, Solari, A, Ortiz, S and Rodríguez, J (2015) Trypanosoma cruzi burden, genotypes, and clinical evaluation of Chilean patients with chronic Chagas cardiopathy. Parasitology Research 114, 30073018.Google Scholar
Aridgides, D, Salvador, R and PereiraPerrin, M (2013) Trypanosoma cruzi highjacks TrkC to enter cardiomyocytes and cardiac fibroblasts while exploiting TrkA for cardioprotection against oxidative stress. Cellular Microbiology 15, 13571366.Google Scholar
Balouz, V, Melli, LJ, Volcovich, R, Moscatelli, G, Moroni, S, González, N, Ballering, G, Bisio, M, Ciocchini, A and Buscaglia, CA (2017) The Trypomastigote Small Surface Antigen from Trypanosoma cruzi improves treatment evaluation and diagnosis in pediatric Chagas disease. Journal of Clinical Microbiology 55, 13171317.Google Scholar
Belaunzaran, LM, Lammel, ME, Gimenez, G, Bott, E, Durante de Isola, EL, Wilkowsky, ES and Barbieri, MA (2013) Phospholipase A(1): a novel virulence factor in Trypanosoma cruzi. Molecular and Biochemical Parasitology 187, 7786.Google Scholar
Bern, C, Kjos, S, Yabseley, M and Montgomery, S (2011) Trypanosoma cruzi and Chagas disease in United States. Clinical Microbiology Reviews 24, 655681.Google Scholar
Bhattacharyya, T, Brooks, J, Yeo, M, Lewis, MD, Llewellyn, MS, Miles, MA and Carrasco, HJ (2010) Analysis of molecular diversity of the Trypanosoma cruzi trypomastigote small surface antigen reveals novel epitopes, evidence of positive selection and potential implications for lineage-specific serology. International Journal for Parasitology 40, 921928.Google Scholar
Bhattacharyya, T, Falconar, AK, Luquetti, AO, Costales, JA, Grijalva, MJ, Lewis, MD, Messenger, LA, Tran, TT, Ramirez, JD, Guhl, F, Carrasco, HJ, Diosque, P, Garcia, L, Litvinov, SV and Miles, MA (2014) Development of peptide-based lineage-specific serology for chronic Chagas disease: geographical and clinical distribution of epitope recognition. PLoS Neglected Tropical Diseases 8, 112.Google Scholar
Branquinha, M, Marinho, F, Sangenito, L, Oliveira, S, Goncalves, K, Ennes-Vidal, V, d'Avila-Levy, C and Santos, A (2013) Calpains: potential targets for alternative chemotherapeutic intervention against human pathogenic trypanosomatids. Current Medicinal Chemistry 20, 31743185.Google Scholar
Brenière, S, Barnabé, C and Waleckx, E (2016) Over six thousand Trypanosoma cruzi strains classified into discrete typing units (DTUs): attempt at an inventory. PLoS Neglected Tropical Diseases 10, 119.Google Scholar
Brown, S, Cornforth, D and Mideo, N (2012) Evolution of virulence in opportunistic pathogens: generalism, plasticity, and control. Trends in Microbiology 20, 336342.Google Scholar
Burgos, JM, Bisio, M, Duffy, T, Levin, M, Schijman, A, Altcheh, J, Burgos Freilij, H, Valadares, H, Freitas, J, Macedo, A, Seidenstein, M, Macchi, L and Piccinali, R (2007) Direct molecular profiling of minicircle signatures and lineages of Trypanosoma cruzi bloodstream populations causing congenital Chagas disease. International Journal for Parasitology 37, 13191327.Google Scholar
Burgos, JM, Diez, M, Vigliano, C, Bisio, M, Risso, M, Duffy, T, Cura, C, Brusses, B, Favaloro, L, Leguizamon, MS, Lucero, RH, Laguens, R, Levin, MJ, Favaloro, R and Schijman, AG (2010) Molecular identification of Trypanosoma cruzi discrete typing units in end-stage chronic Chagas heart disease and reactivation after heart transplantation. Clinical Infectious Diseases 51, 485495.Google Scholar
Burleigh, BA and Woolsey, AM (2002) Cell signalling and Trypanosoma cruzi invasion. Cellular Microbiology 4, 701711.Google Scholar
Burleigh, BA, Caler, E, Webster, P and Andrews, N (1997) A cytosolic serine endopeptidase from Trypanosoma cruzi is required for the generation of Ca2+ signaling in mammalian cells. Journal of Cell Biology 13, 609620.Google Scholar
Cámara, M, Cánepa, GE, Lantos, AB, Balouz, V, Yu, H, Chen, X, Campetella, O, Mucci, J and Buscaglia, CA (2017) The trypomastigote small surface antigen (TSSA) regulates Trypanosoma cruzi infectivity and differentiation. PLoS Neglected Tropical Diseases 11, 121.Google Scholar
Camargo, R, Faria, LO, Kloss, A, Favali, CBF, Kuckelkorn, U, Kloetzel, PM, de Sá, CM and Lima, BD (2014) Trypanosoma cruzi infection down-modulates the immunoproteasome biosynthesis and the MHC class I cell surface expression in HeLa cells. PLoS ONE 9, 112.Google Scholar
Carareto, C, Manoel-Caetano, FS, Aparecida, CM, Silva, AE, Borim, AA and Miyazaki, K (2008) kDNA gene signatures of Trypanosoma cruzi in blood and oesophageal mucosa from chronic chagasic patients. Transactions of the Royal Society of Tropical Medicine and Hygiene 102, 11021107.Google Scholar
Carranza, JC, Valadares, HMS, D’Ávila, DA, Baptista, RP, Moreno, M, Galvão, LMC, Chiari, E, Sturm, NR, Gontijo, ED, Macedo, AM and Zingales, B (2009) Trypanosoma cruzi maxicircle heterogeneity in Chagas disease patients from Brazil. International Journal for Parasitology 39, 963973.Google Scholar
Carrasco, HJ, Segovia, M, Llewellyn, MS, Morocoima, A, Urdaneta-Morales, S, Martínez, C, Garcia, C, Rodríguez, M, Espinosa, R, de Noya, BA, Díaz-Bello, Z, Herrera, L, Fitzpatrick, S, Yeo, M, Miles, MA and Feliciangeli, MD (2012) Geographical distribution of Trypanosoma cruzi genotypes in Venezuela. PLoS Neglected Tropical Diseases 6, 19.Google Scholar
Chuenkova, MV and PereiraPerrin, M (2009) Trypanosoma cruzi targets Akt in host cells as an intracellular antiapoptotic strategy. Science Signaling 2, 120.Google Scholar
Chuenkova, MV and PereiraPerrin, M (2011) Neurodegeneration and neuroregeneration in Chagas disease. Advances in Parasitology 76, 195233.Google Scholar
Coates, BM, Sullivan, DP, Makanji, MY, Du, NY, Olson, CL, Muller, WA, Engman, DM and Epting, CL (2013) Endothelial transmigration by Trypanosoma cruzi. PLoS ONE 8, 110.Google Scholar
Colorado, IA, Acquatella, H, Catalioti, F, Fernandez, MT and Layrisse, Z (2000) Original articles: HLA class II DRB1, DQB1, DPB1 polymorphism and cardiomyopathy due to Trypanosoma cruzi chronic infection. Human Immunology 61, 320325.Google Scholar
Corral, RS, Guerrero, NA, Cuervo, H, Girones, N and Fresno, M (2013) Trypanosoma cruzi infection and endothelin-1 cooperatively activate pathogenic inflammatory pathways in cardiomyocytes. PLoS Neglected Tropical Diseases 7, 112.Google Scholar
Costa, G, Moreira, PR, Menezes, CAS, Silva, M, Dutra, WO, Rocha, MO and Gollob, KJ (2009) Functional IL-10 gene polymorphism is associated with Chagas disease cardiomyopathy. Journal of Infectious Diseases 199, 451454.Google Scholar
Cruz, MC, Souza-Melo, N, da Silva, CV, DaRocha, WD, Bahia, D, Araújo, PR, Teixeira, SR and Mortara, RA (2012) Trypanosoma cruzi: role of δ-amastin on extracellular amastigote cell invasion and differentiation. PLoS ONE 7, 111.Google Scholar
Cruz, J, Santos-Miranda, A, Monti-Rocha, R, Machado, F, Sales, P, Campos, P and Roman-Campos, D (2016) Altered cardiomyocyte function and Trypanosoma cruzi persistence in Chagas disease. American Journal of Tropical Medicine and Hygiene 94, 10281033.Google Scholar
Cunha-Neto, E and Chevillard, C (2014) Chagas disease cardiomyopathy: immunopathology and genetics. Mediators of Inflammation 2014, 111.Google Scholar
Cupello, MP, Souza, CF, Nogueira, NP, Laranja, GA, Sabino, KC, Coelho, MG, Oliveira, MM and Paes, MC (2014) Trypanosomatid essential metabolic pathway: new approaches about heme fate in Trypanosoma cruzi. Biochemical and Biophysical Research Communications 449, 216221.Google Scholar
Cura, C, Bisio, M, Duffy, T, Schijman, A, Lucero, RH, Formichelli, LB, Brusés, BL, Merino, DE, Oshiro, E, Sosa-Estani, S, Burgos, J, Lejona, S, Anchart, E, Hernández, DO, Severini, GV, Velazquez, E, Lattes, R, Altcheh, J, Freilij, H, Diez, M, Nagel, C, Vigliano, C, Favaloro, L and Favaloro, RR (2012) Trypanosoma cruzi discrete typing units in Chagas disease patients from endemic and non-endemic regions of Argentina. Parasitology 139, 516521.Google Scholar
Cura, CI, Duffy, T, Lucero, RH, Bisio, M, Péneau, J, Jimenez-Coello, M, Calabuig, E, Gimenez, MJ, Valencia, E, Kjos, SA, Santalla, J, Mahaney, SM, Cayo, NM, Nagel, C, Barcán, L, Málaga Machaca, ES, Acosta, KY, Brutus, L, Ocampo, SB and Aznar, C (2015) Multiplex real-time PCR assay using TaqMan probes for the identification of Trypanosoma cruzi DTUs in biological and clinical samples. PLoS Neglected Tropical Diseases 9, 118.Google Scholar
Dario, M, Rodrigues, M, Barros, J, Xavier, S, Roque, A, Jansen, A and D'Andrea, P (2016) Ecological scenario and Trypanosoma cruzi DTU characterization of a fatal acute Chagas disease case transmitted orally (Espírito Santo state, Brazil). Parasites and Vectors 9, 114.Google Scholar
de Souza, W, de Carvalho, TMU and Barrias, ES (2010) Review on Trypanosoma cruzi: host cell interaction. International Journal of Cell Biology 1, 118.Google Scholar
Deghaide, NHS, Dantas, RO and Donadi, EA (1998) HLA class I and II profiles of patients presenting with Chagas’ disease. Digestive Diseases and Sciences 43, 246252.Google Scholar
del Puerto, R, Nishizawa, JE, Kikuchi, M, Iihoshi, N, Roca, Y, Avilas, C, Gianella, A, Lora, J, Velarde, F, Gutierrez, U, Renjel, LA, Miura, S, Higo, H, Komiya, N, Maemura, K and Hirayama, K (2010) Lineage analysis of circulating Trypanosoma cruzi parasites and their association with clinical forms of Chagas disease in Bolivia. PLoS Neglected Tropical Diseases 4. doi: 10.1371/journal.pntd.0000687.Google Scholar
Dias, JC, Ramos, AN, Gontijo, ED, Luquetti, A, Shikanai-Yasuda, MA, Coura, JR, Torres, RM, Melo, JR, Almeida, EA, Oliveira, WJ, Silveira, AC, Rezende, JM, Pinto, FS, Ferreira, AW, Rassi, A, Fragata, AA Filho, Sousa, AS, Correia, D, Jansen, AM, Andrade, GM, Britto, CF, Pinto, AY, Rassi, AJ, Campos, DE, Abad-Franch, F, Santos, SE, Chiari, E, Hasslocher-Moreno, AM, Moreira, EF, Marques, DS, Silva, EL, Marin-Neto, JA, Galvão, LM, Xavier, SS, Valente, SA, Carvalho, NB, Cardoso, AV, Silva, RA, Costa, VM, Vivaldini, SM, Oliveira, SM, Valente, VD, Lima, MM and Alves, RV (2016). 2nd Brazilian consensus on Chagas disease, 2015. Revista da Sociedade Brasileira de Medicina Tropical 49, 359.Google Scholar
Díaz, ML, Torres, R and González, CI (2011) Differential protein expression in developmental stages of Trypanosoma cruzi I isolated from a patient with chronic chagasic cardiomyopathy. Biomédica 31, 503513.Google Scholar
Drigo, SA, Cunha-Neto, E, Ianni, B, Faé, KC, Nunes, VL, Buck, P, Mady, C, Kalil, J, Goldberg, AC, Cardoso, MRA and Braga, PE (2006) TNF gene polymorphisms are associated with reduced survival in severe Chagas’ disease cardiomyopathy patients. Microbes and Infection 8, 598603.Google Scholar
Duffy, T, Bisio, M, Altcheh, J, Burgos, JM, Diez, M, Levin, MJ, Favaloro, RR, Freilij, H and Schijman, AG (2009) Accurate real-time PCR strategy for monitoring bloodstream parasitic loads in Chagas disease patients. PLoS Neglected Tropical Diseases 3, 110.Google Scholar
El-Sayed, NM, Bartholomeu, DC, Ghedin, E, Delcher, AL, Blandin, G, Westenberger, SJ, Caler, E, Cerqueira, GC, Haas, B, Crabtree, J, Feldblyum, T, Hou, L, Koo, H, Lacerda, D, Pai, G, Pop, M, Salzberg, SL, Shetty, J, Simpson, AJ, Van Aken, S, Wortman, J, White, O, Fraser, CM, Myler, PJ, Aggarwal, G, Worthey, EA, Anupama, A, Attipoe, P, Cadag, E, Fazelina, G, Huang, Y, Louie, T, Nelson, S, Parsons, M, Pentony, M, Rinta, J, Robertson, L, Seyler, A, Sisk, E, Vogt, C, Stuart, KD, Nilsson, D, Tran, AN, Branche, C, Arner, E, Bontempi, E, Darban, H, Edwards, K, Ferella, M, Kindlund, E, Kluge, S, McKenna, A, Mizuno, Y, Ochaya, S, Tammi, MT, Andersson, B, Campbell, DA, Machado, CR, Teixeira, S, Åslund, L, Pettersson, U, Bringaud, F, Burton, P, McCulloch, R, Mottram, JC, Ward, PN, Carrington, M, Sharma, R, Da Silveira, JF, De Jong, P, Osoegawa, K, Englund, PT, Frasch, AC, Sanchez, DO, Gull, K, Wickstead, B, Horn, D, Klingbeil, M, Levin, MJ, Lorenzi, H, Ramirez, JL and Tarleton, R (2005) The genome sequence of Trypanosoma cruzi, etiologic agent of chagas disease. Science 309, 409415.Google Scholar
Esper, L, Utsch, L, Brant, F, Teixeira, MM, Vieira, LQ, Machado, FS, Soriani, FM, Arantes, RM, Campos, CF, Pinho, V, Souza, DG and Tanowitz, HB (2014) Regulatory effects of IL-18 on cytokine profiles and development of myocarditis during Trypanosoma cruzi infection. Microbes and Infection 16, 481490.Google Scholar
Fernández-Mestre, MT, Montagnani, S and Layrisse, Z (2004) Original article: is the CCR5-59029-G/G genotype a protective factor for cardiomyopathy in Chagas disease? Human Immunology 65, 725728.Google Scholar
Ferreira, ÉC, Portocarrero, AR, Brustolin, CF, Ciupa, L, Massini, PF, Aleixo, DL and de Araújo, SM (2017) Phosphorus protects cardiac tissue by modifying the immune response in rats infected by Trypanosoma cruzi. Cytokine 17, 102106.Google Scholar
Flórez, O, Zafra, G, Morillo, C, Martín, J and González, CI (2006) Interleukin-1 gene cluster polymorphism in chagas disease in a Colombian case-control study. Human Immunology 67, 741748.Google Scholar
Franzén, O, Ochaya, S, Sherwood, E, Andersson, B, Lewis, MD, Llewellyn, MS and Miles, MA (2011) Shotgun sequencing analysis of Trypanosoma cruzi i Sylvio X10/1 and comparison with T. cruzi VI CL Brener. PLoS Neglected Tropical Diseases 5, 19.Google Scholar
Freire-de-Lima, L, Fonseca, LM, Oeltmann, T, Mendonça-Previato, L and Previato, JO (2015) The trans-sialidase, the major Trypanosoma cruzi virulence factor: Three decades of studies. Glycobiology 25, 11421149.Google Scholar
Gaunt, MW, Yeo, M, Frame, IA, Stothard, JR, Carrasco, HJ, Taylor, MC, Mena, S, Solis, P, Miles, GAJ, Acosta, N, de Arias, AR and Miles, MA (2003) Mechanism of genetic exchange in American trypanosomes. Nature 421, 936939.Google Scholar
Guedes, PMM, Gutierrez, FRS, Silva, GK, Dellalibera-Joviliano, R, Rodrigues, GJ, Bendhack, LM, Rassi, A, Schmidt, A, Maciel, BC, Marin- Neto, JA and Silva, JS (2012) Deficient regulatory T cell activity and low frequency of IL-17-producing T cells correlate with the extent of cardiomyopathy in human Chagas’ disease. PLoS Neglected Tropical Diseases 6, e1630.Google Scholar
Guhl, F (2013). Epidemiologia molecular de Trypanosoma cruzi 1:1, 1-8. Retrieved from Redalyc website. Available at http://www.redalyc.org/articulo.oa?id=17027695001.Google Scholar
Guhl, F and Ramírez, JD (2013) Retrospective molecular integrated epidemiology of Chagas disease in Colombia. Infection, Genetics and Evolution 20, 148154.Google Scholar
Guhl, F, Auderheide, A and Ramírez, JD (2014) From ancient to contemporary molecular eco-epidemiology of Chagas disease in the Americas. International Journal for Parasitology 44, 605612.Google Scholar
Hassan, G, Mukherjee, S, Nagajyothi, F, Weiss, L and Petkova, S (2006) Trypanosoma cruzi infection induces proliferation of vascular smooth muscle cells. Infection and Immunity 74, 152159.Google Scholar
Henrique, PM, Marques, T, da Silva, MV, de Oliveira, CF, Rodrigues, V, Gomez-Hernandez, C, Ramirez, LE, Ferreira, WS, Nogueira, GA and Norris, KA (2016) Correlation between the virulence of T. cruzi strains, complement regulatory protein expression levels, and the ability to elicit lytic antibody production. Experimental Parasitology 170, 6672.Google Scholar
Hernández, C, Cucunubá, Z, Parra, E, Toro, G, Zambrano, P and Ramírez, JD (2014) Chagas disease (Trypanosoma cruzi) and HIV co-infection in Colombia. International Journal of Infectious Diseases 26, 146148.Google Scholar
Hernández, C, Cucunubá, Z, Flórez, C, Olivera, M, Valencia, C, Zambrano, P, León, C and Ramírez, JD (2016) Molecular diagnosis of chagas disease in Colombia: parasitic loads and discrete typing units in patients from acute and chronic phases. PLoS Neglected Tropical Diseases 10, e0004997.Google Scholar
Higuera, SL, Guhl, F and Ramírez, JD (2013) Identification of Trypanosoma cruzi discrete typing units (DTUs) through the implementation of a high-resolution melting (HRM) genotyping assay. Parasites & Vectors 6, 16.Google Scholar
Jackson, AP (2010) The evolution of amastin surface glycoproteins in Trypanosomatid parasites. Molecular Biology and Evolution 27, 3345.Google Scholar
Jackson, Y, Pinto, A and Pett, S (2014) Chagas disease in Australia and New Zealand: risks and needs for public health interventions. Tropical Medicine & International Health 19, 212218.Google Scholar
Kuete, V (2013) Medicinal Plant Research in Africa: Pharmacology and Chemistry, 1st Edn. London, UK: Elsevier Insights.Google Scholar
Lasso, P, Beltrán, L, Guzmán, F, Rosas, F, Thomas, MC, López, MC, González, JM, Cuéllar, A and Puerta, C (2016) Promiscuous recognition of a Trypanosoma cruzi CD8+ T cell epitope among HLA-A2, HLA-A24 and HLA-A1 supertypes in chagasic patients. PLoS ONE 11, e0150996.Google Scholar
Lauthier, JJ, Tomasini, N, Rumi, MM, D'Amato, AMA, Ragone, PG, Diosque, P, Barnabé, C, Tibayrenc, M, Yeo, M, Lewis, MD, Llewellyn, MS, Miles, MA and Basombrío, MA (2012) Candidate targets for multilocus sequence typing of Trypanosoma cruzi: validation using parasite stocks from the Chaco Region and a set of reference strains. Infection, Genetics and Evolution 12, 350358.Google Scholar
Layrisse, Z, Fernandez, MT, Montagnani, S, Matos, M, Balbas, O, Herrera, F, Colorado, IA, Catalioti, F and Acquatella, H (2000) HLA-C*03 is a risk factor for cardiomyopathy in Chagas disease. Human Immunology 61, 925929.Google Scholar
Leiby, DA, Nguyen, ML, Proctor, MC, Townsend, RL and Stramer, SL (2017) Frequency of Trypanosoma cruzi parasitemia among infected blood donors with a potential association between parasite lineage and transfusion transmission. Transfusion 57, 14261432.Google Scholar
Leon, CM, Ramirez, JD, Montilla, M, Vanegas, R, Castillo, M and Parra, E (2017) Murine models susceptibility to distinct Trypanosoma cruzi I genotypes infection. Parasitology 144, 512519.Google Scholar
Leon Rodrigez, DA, Carmona, FD, Echeverría, LE, González, CI and Martin, J (2016) IL18 gene variants influence the susceptibility to chagas disease. PLoS Neglected Tropical Diseases 10, e0004583.Google Scholar
Lewis, MD, Yeo, M, Llewellyn, MS, Miles, MA, Ma, J and Carrasco, HJ (2009) Genotyping of Trypanosoma cruzi: systematic selection of assays allowing rapid and accurate discrimination of all known lineages. American Journal of Tropical Medicine and Hygiene 81, 10411049.Google Scholar
Lima, L, Espinosa-Álvarez, O, Ortiz, PA, Camargo, EP, Teixeira, MMG, Trejo-Varón, JA, Carranza, JC, Pinto, CM, Serrano, MG and Buck, GA (2015 a) Genetic diversity of Trypanosoma cruzi in bats, and multilocus phylogenetic and phylogeographical analyses supporting Tcbat as an independent DTU (discrete typing unit). Acta Tropica 151, 166177.Google Scholar
Lima, L, Espinosa-Álvarez, O, Pinto, CM, Cavazzana, M Jr, Pavan, AC, Carranza, JC, Lim, BK, Campaner, M, Takata, CSA, Camargo, EP, Hamilton, PB and Teixeira, MMG (2015 b) New insights into the evolution of the Trypanosoma cruzi clade provided by a new trypanosome species tightly linked to Neotropical Pteronotus bats and related to an Australian lineage of trypanosomes. Parasites & Vectors 8, 118.Google Scholar
Llewellyn, MS, Miles, MA, Carrasco, HJ, Lewis, MD, Yeo, M, Vargas, J, Torrico, F, Diosque, P, Valente, V, Valente, SA and Gaunt, MW (2009) Genome-scale multilocus microsatellite typing of Trypanosoma cruzi discrete typing unit I reveals phylogeographic structure and specific genotypes linked to human infection. PLoS Pathogens 5, e1000410.Google Scholar
Llewellyn, MS, Messenger, LA, Luquetti, AO, Garcia, L, Torrico, F, Tavares, SBN, Cheaib, B, Derome, N, Delepine, M, Baulard, C, Deleuze, JF, Sauer, S, Miles, and Michael, A (2015) Deep sequencing of the Trypanosoma cruzi GP63 surface proteases reveals diversity and diversifying selection among chronic and congenital Chagas disease patients. PLoS Neglected Tropical Diseases 9, 123.Google Scholar
Longhi, SA, Tasso, LM, Gomez, KA, Atienza, A, Bonato, R, Chiale, P, Perez, G, Buying, A, Pinilla, C, Judkowski, VA, Balouz, V, Buscaglia, CA and Santos, R (2014) Cytokine production but lack of proliferation in peripheral blood mononuclear cells from chronic Chagas’ disease cardiomyopathy patients in response to T. cruzi ribosomal P proteins. PLoS Neglected Tropical Diseases 8, e2906.Google Scholar
Luquetti, AO, Tavares, S, Siriano, L, Oliveira, R, Campos, DE, Morais, CA and Oliveira, EC (2015) Congenital transmission of Trypanosoma cruzi in central Brazil. A study of 1211 individuals born to infected mothers. Memórias do Instituto Oswaldo Cruz 110, 369376.Google Scholar
Macedo, AM and Segatto, M (2010). Implications of Trypanosoma Cruzi Intraspecific Diversity in the Pathogenesis of Chagas Disease, in American Trypanosomiasis Chagas Disease One Hundred Years of Research, 2nd Edn. Montpellier, France: Academic Press.Google Scholar
Macedo, AM, Machado, CR, Oliveira, RP and Pena, SDJ (2004) Trypanosoma cruzi: genetic structure of populations and relevance of genetic variability to the pathogenesis of Chagas disease. Memórias do Instituto Oswaldo Cruz 99, 112.Google Scholar
Magalhães, LM, Villani, FN, Nunes, MC, Gollob, KJ, Rocha, MO and Dutra, WO (2013) High interleukin 17 expression is correlated with better cardiac function in human Chagas disease. The Journal of Infectious Diseases 207, 661665.Google Scholar
Malvezi, AD, Da Silva, RV, De Freitas, RC, Lovo-Martins, MI, Tatakihara, VLH, Zanluqui, NG, Neto, EC, Goldenberg, S, Bordignon, J, Yamada-Ogatta, SF, Martin-Pinge, MC, Cecchini, R and Pinge-Filho, P (2014) Inhibition of cyclooxygenase-1 and cyclooxygenase-2 impairs Trypanosoma cruzi entry into cardiac cells and promotes differential modulation of the inflammatory response. Antimicrobial Agents and Chemotherapy 58, 61576164.Google Scholar
Mantilla, JC, Zafra, GA, Macedo, AM and González, CI (2010) Mixed infection of Trypanosoma cruzi I and II in a Colombian cardiomyopathic patient. Human Pathology 4, 610613.Google Scholar
Marin-Neto, AJ, Rassi, A Jr and Maciel, BC (2015). Chagas disease: Pathology and pathogenesis. UpToDate. Avalaible at https://www.uptodate.com/contents/chagas-disease-pathology-and-pathogenesis.Google Scholar
Margioto Teston, AP, de Abreu, AP, Abegg, CP, Gomes, ML and de Ornelas Toledo, MJ (2017) Outcome of oral infection in mice inoculated with Trypanosoma cruzi IV of the Western Brazilian Amazon. Acta Tropica. 166, 212217.Google Scholar
Meloni, M, Caporali, A, Graiani, G, Lagrasta, C, Katare, R, Van Linthout, S, Spillmann, F, Campesi, I, Madeddu, P, Quaini, F and Emanueli, C (2010) Nerve growth factor promotes cardiac repair following myocardial infarction. Circulation Research 106, 1275–U1230.Google Scholar
Messenger, LA, Llewellyn, MS, Bhattacharyya, T, Franzén, O, Lewis, MD, Ramírez, JD, Carrasco, HJ, Andersson, B and Miles, MA (2012) Multiple mitochondrial introgression events and heteroplasmy in Trypanosoma cruzi revealed by maxicircle MLST and next generation sequencing. PLoS Neglected Tropical Diseases 6, 112.Google Scholar
Messenger, LA, Ramirez, JD, Llewellyn, MS, Guhl, F and Miles, MA (2016) Importation of hybrid human-associated Trypanosoma cruzi strains of southern South American origin, Colombia. Emerging Infectious Diseases 22, 14521455.Google Scholar
Miles, MA, Póvoa, M, Prata, A, Cedillos, RA, De Souza, AA and Macedo, M (1981) Do radically dissimilar Trypanosoma cruzi strains (zymodemes) cause Venezuelan and Brazilian forms of chagas’ disease? The Lancet 317, 13381340.Google Scholar
Moncayo, A and Yanine, MIO (2006) An update on Chagas disease (human American trypanosomiasis). Annals of Tropical Medicine & Parasitology 100, 663677.Google Scholar
Monteiro, FA, Peretolchina, T, Lazoski, C, Harris, K, Dotson, EM, Abad-Franch, F, Tamayo, E, Pennington, PM, Monroy, C, Cordon-Rosales, C, Salazar-Schettino, PM, Gómez-Palacio, A, Grijalva, MJ, Beard, CB and Marcet, PL (2013a) Phylogeographic pattern and extensive mitochondrial DNA divergence disclose a species complex within the Chagas disease vector Triatoma dimidiata. PLoS ONE 8, 115.Google Scholar
Monteiro, WM, Margioto-Teston, AP, Gruendling, AP, dos Reis, D, Gomes, ML, de Araújo, SM, Bahia, MT, Magalhães, LK, de Oliveira-Guerra, JA, Silveira, H, Toledo, MJ and Vale-Barbosa, M (2013b). Trypanosoma cruzi I and IV stocks from Brazilian Amazon are divergent in terms of biological and medical properties in mice. PLoS Neglected Tropical Diseases 7. doi: 10.1371/journal.pntd.0002069.Google Scholar
Nagib, PRA, Dutra, WO, Chiari, E, Conceição, RS and Machado, CRS (2007) Trypanosoma cruzi: populations bearing opposite virulence induce differential expansion of circulating CD3+CD4−CD8− T cells and cytokine serum levels in young and adult rats. Experimental Parasitology 116, 366374.Google Scholar
Nogueira, LG, Santos, RH, Ianni, BMFiorelli, AI, Mairena, EC, Benvenuti, LA, Frade, , Donadi, E, Dias, F, Saba, B, Wang, HT, Fragata, A, Sampaio, M, Hirata, MH, Buck, P, Mady, C, Bocchi, EA, Stolf, NA, Kalil, J and Cunha-Neto, E (2012). Myocardial chemokine expression and intensity of myocarditis in Chagas cardiomyopathy are controlled by polymorphisms in CXCL9 and CXCL10. PLoS Neglected Tropical Diseases 6, 113.Google Scholar
Nogueira, LG, Frade, AF, Ianni, BM, Laugier, L, Pissetti, CW, Cabantous, S, Baron, M, Peixoto-Gde, L, Borges-Ade, M, Donadi, E, Marin-Neto, JA, Schmidt, A, Dias, F, Saba, B, Wang, HT, Fragata, A, Sampaio, M, Hirata, MH, Buck, P, Mady, C, Martinelli, M, Lensi, M, Siqueira, SF, Pereira, AC, Rodrigues, VJ, Kalil, J, Chevillard, C and Cunha-Neto, E (2015) Functional IL18 polymorphism and susceptibility to Chronic Chagas Disease. Cytokine 73, 7983.Google Scholar
Okura, M, Fang, J, Salto, ML, Singer, RS, Docampo, R and Moreno, SN (2005) A lipid-modified phosphoinositide-specific phospholipase C (TcPI-PLC) is involved in differentiation of trypomastigotes to amastigotes of Trypanosoma cruzi. Journal of Biological Chemistry 280, 1623516243.Google Scholar
Osorio, L, Rios, I, Gutierrez, B and Gonzalez, J (2012) Virulence factors of Trypanosoma cruzi: who is who? Microbes and Infection 14, 13901402.Google Scholar
Petkova, SB, Tanowitz, HB, Magazine, HI, Factor, SM, Chan, J, Pestell, RG, Bouzahzah, B, Douglas, SA, Shtutin, V, Morris, SA, Tsang, E, Weiss, LM, Christ, GJ, Wittner, M and Huang, H (2000) Myocardial expression of endothelin-1 in murine Trypanosoma cruzi infection. Cardiovascular Pathology 9, 257265.Google Scholar
Pinto, CM, Kalko, EK, Cottontail, VM, Cottontail, I and Wellinghausen, N (2012) Tcbat a bat-exclusive lineage of Trypanosoma cruzi in the Panama Canal Zone, with comments on its classification and the use of the 18S rRNA gene for lineage identification. Infection, Genetics and Evolution 12, 13281332.Google Scholar
Pinto, CM, Ocaña-Mayorga, S, Tapia, E, Lobos, S, Zurita, A, Aguirre-Villacís, , MacDonals, A, Villacis, AG, Lima, L, Teixeira, MM, Grijalva, MJ and Perkins, S (2015) Bats, trypanosomes, and triatomines in Ecuador: new insights into the diversity, transmission, and origins of Trypanosoma cruzi and Chagas disease. PLoS ONE 10, e0139999.Google Scholar
Pissetti, CW, Correia, D, de Oliveira, RF, Llaguno, MM, Balarin, MA, Silva-Grecco, RL and Rodrigues, JV (2011) Genetic and functional role of TNF-alpha in the development Trypanosoma cruzi infection. PLoS Neglected Tropical Diseases 5, 110.Google Scholar
Poveda, C, Fresno, M, Gironès, NA, Martins-Filho, O, Ramírez, JD, Santi-Rocca, J, Marín-Neto, JA, Morillo, CA, Rosas, and Guhl, F (2014) Cytokine profiling in Chagas disease: towards understanding the association with infecting Trypanosoma cruzi discrete typing units (a BENEFIT TRIAL sub-study). PLoS ONE 9.Google Scholar
Prata, A (2001) Clinical and epidemiological aspects of Chagas disease. Lancet Infectious Diseases 1, 92100.Google Scholar
Ramasawmy, R, Faé, KC, Cunha-Neto, E, Borba, SC, Ianni, B, Mady, C, Goldberg, AC and Kalil, J (2008) Variants in the promoter region of IKBL/NFKBIL1 gene may mark susceptibility to the development of chronic Chagas’ cardiomyopathy among Trypanosoma cruzi-infected individuals. Molecular Immunology 45, 283288.Google Scholar
Ramírez, JD and Hernández, C (2017) Trypanosoma cruzi I: towards the need of genetic subdivision?, Part II. Acta Tropica S0001-706X, 3025030254.Google Scholar
Ramírez, JD and Llewellyn, MS (2014) Reproductive clonality in protozoan pathogens-truth or artefact? Molecular Ecology 23, 41954202.Google Scholar
Ramírez, JD, Guhl, F, Umezawa, ES, Morillo, CA, Rosas, F, Marin-Neto, JA and Restrepo, S (2009) Evaluation of adult chronic Chagas’ heart disease diagnosis by molecular and serological methods. Journal of Clinical Microbiology 47, 39453951.Google Scholar
Ramírez, JD, Guhl, F, Rendón, LM, Rosas, F, Marin-Neto, JA and Morillo, C (2010) Chagas cardiomyopathy manifestations and Trypanosoma cruzi genotypes circulating in chronic chagasic patients. PLoS Neglected Tropical Diseases 4.Google Scholar
Ramírez, JD, Guhl, F, Messenger, LA, Lewis, MD, Montilla, M, Cucunuba, Z and Llewellyn, MS (2012) Contemporary cryptic sexuality in Trypanosoma cruzi. Molecular Ecology 21, 42164226.Google Scholar
Ramírez, JD, Montilla, M, Cucunubá, ZM, Floréz, AC, Zambrano, P and Guhl, F (2013) Molecular epidemiology of human oral chagas disease outbreaks in Colombia. PLoS Neglected Tropical Diseases 7, 17.Google Scholar
Ramírez, JD, Hernández, C, Cucunubá, ZM, Montilla, M, Flórez, AC, Zambrano, P and Parra, E (2014) First report of human Trypanosoma cruzi infection attributed to TcBat genotype. Zoonoses and Public Health 61, 477479.Google Scholar
Rassi, JA, Rassi, A and Marin-Neto, JA (2009) Chagas heart disease: pathophysiologic mechanisms, prognostic factors and risk stratification. Memórias do Instituto Oswaldo Cruz 104, 152158.Google Scholar
Rassi, JA, Rassi, A and Marin-Neto, JA (2010) Chagas disease. The Lancet 375, 13881402.Google Scholar
Rassi, JA, Rassi, A and Marcondes de Rezende, J (2012) American trypanosomiasis (Chagas disease). Infectious Disease Clinics of North America 2, 275291.Google Scholar
Rassi, JA, Rassi, A and Marin-Neto, JA (2015) Chagas disease. The Lancet 375, 4571.Google Scholar
Rassi, JA, Marin Neto, JA and Rassi, A (2017) Chronic Chagas cardiomyopathy: a review of the main pathogenic mechanisms and the efficacy of aetiological treatment following the BENznidazole Evaluation for Interrupting Trypanosomiasis (BENEFIT) trial. Memórias do Instituto Oswaldo Cruz 112, 224235.Google Scholar
Reis, DD, Jones, EM, Tostes, S Jr, Lopes, ER, Gazzinelli, G, Colley, DG and McCurley, TL (1993). Characterization of inflammatory infiltrates in chronic chagasic myocardial lesions: presence of tumor necrosis factor-alpha+ cells and dominance of granzyme A+, CD8+ lymphocytes. The American Journal of Tropical Medicine and Hygiene 48, 637644.Google Scholar
Reis, PG, Sakita, KM, de Moraes, AG, Aquino, JS, Macedo, LC, Mazini, PS, Sell, AM, de Oliveira, DS, Bulgarelli, R and Laguila, JE (2017) Genetic polymorphisms of IL17 and Chagas disease in the south and southeast of Brazil. Journal of Immunology Research 2017, 7.Google Scholar
Rodrigues, MM, Oliveira, AC and Bellio, M (2012) The immune response to Trypanosoma cruzi : role of toll-like receptors and perspectives for vaccine development. Journal of Parasitology Research 2012, 112.Google Scholar
Rodríguez, JA, Marigorta, UM and Navarro, A (2014) Integrating genomics into evolutionary medicine. Current Opinion in Genetics & Development 29, 97102.Google Scholar
Roggero, E, Rosa Perez, A, Pollachini, N, Raquel Villar, S, Wildmann, J, Besedovsky, H and del Rey, A (2016) The sympathetic nervous system affects the susceptibility and course of Trypanosoma cruzi infection. Brain Behavior and Immunity 58, 228236.Google Scholar
Rojas, MW, Anaya, JM, Gómez, LM, Aristizabal, BH, Cano, R, LE and Lopera, HD (2017) Inmunología de Rojas (W. R. M. Ed. 18 ed.). Medellin (Antioquia, Colombia): Corporación para Investigaciones Biologicas. CIB 2017.Google Scholar
Rozas, M, Doncker, SD, Adaui, V, Coronado, X, Barnabé, C, Tibyarenc, M and Dujardin, JC (2007) Multilocus polymerase chain reaction restriction fragment-length polymorphism genotyping of Trypanosoma cruzi (Chagas disease): taxonomic and clinical applications. The Journal of Infectious Diseases 195, 1381.Google Scholar
Salomone, O, Caeiro, T, Madoery, R, Amuchastegui, M, Omelinauk, M, Juri, D and Kaski, J (2001) High plasma immunoreactive endothelin levels in patients with Chagas’ cardiomyopathy. American Journal of Cardiology 87, 12171220.Google Scholar
San Francisco, J, Gutierrez, B, Neira, I, Munoz, C, Sagua, H, Araya, JE, Andrade, JC, Zailberger, A, Catalán, A, Remonsellez, F, Vega, JL and González, J (2017) Decreased cruzipain and gp85/trans-sialidase family protein expression contributes to loss of Trypanosoma cruzi trypomastigote virulence. Microbes and Infection 19, 5561.Google Scholar
Sanjabi, S, Zenewicz, LA, Kamanaka, M and Flavell, RA (2009) Anti-inflammatory and pro-inflammatory roles of TGF-β, IL-10, and IL-22 in immunity and autoimmunity. Current Opinion in Pharmacology 9, 447453.Google Scholar
Sanmarco, LM, Visconti, LM, Eberhardt, N, Ramello, MC, Ponce, NE, Spitale, NB, Vozza, ML, Bernardi, GA, Gea, S, Minguez, AR and Aoki, MP (2016) IL-6 improves the nitric oxide-induced cytotoxic CD8+ T cell dysfunction in human chagas disease. Frontiers in Immunology 7, 112.Google Scholar
Sanoja, C, Carbajosa, S, Fresno, M and Gironès, N (2013) Analysis of the dynamics of infiltrating CD4+ T cell subsets in the heart during experimental Trypanosoma cruzi infection. PLoS ONE 8, 111.Google Scholar
Santi-Rocca, J, Fernandez-Cortes, F, Chillon-Marinas, C, Gonzalez-Rubio, ML, Girones, N, Fresno, M and Martin, D (2017) A multi-parametric analysis of Trypanosoma cruzi infection: common pathophysiologic patterns beyond extreme heterogeneity of host responses. Scientific Reports 7, 12.Google Scholar
Savino, W (2017) Endocrine immunology of chagas disease. In Endocrine Immunology. Karger Publishers, 160175. https://www.karger.com/Article/Abstract/452914Google Scholar
Segovia, M, Carrasco, HJ, Martínez, CE, Messenger, LA, Nessi, A, Londoño, JC, Espinosa, R, Martínez, C, Mijares, A, Bonfante-Cabarcas, R, Lewis, MD, de Noya, BA, Miles, MA and Llewellyn, MS (2013) Molecular epidemiologic source tracking of orally transmitted Chagas disease, Venezuela. Emerging Infectious Diseases 19, 10981101.Google Scholar
Strasen, J, Williams, T, Ertl, G, Ritter, O, Zoller, T and Stich, A (2014) Epidemiology of Chagas disease in Europe: many calculations, little knowledge. Clinical Research in Cardiology 103, 110.Google Scholar
Sturm, NR, Vargas, NS, Westenberger, SJ, Zingales, B and Campbell, DA (2003) Evidence for multiple hybrid groups in Trypanosoma cruzi. International Journal for Parasitology 33, 269279.Google Scholar
Sánchez, LV and Ramírez, JD (2013) Congenital and oral transmission of American trypanosomiasis: an overview of physiopathogenic aspects. Parasitology 140, 147159.Google Scholar
Sánchez-Montalvá, A, Salvador, F, Rodríguez-Palomares, J, Sulleiro, E, Sao-Avilés, A, Roure, S, Valerio, L, Evangelista, A and Molina, I (2016). Chagas cardiomyopathy: usefulness of EKG and echocardiogram in a non-endemic country. PLoS ONE 11. doi: 10.1371/journal.pone.0157597.Google Scholar
Tarleton, RL (1991) Regulation of immunity in Trypanosoma cruzi infection. Experimental Parasitology 73, 106109.Google Scholar
Tarleton, RL (2003). Chagas disease: a role for autoimmunity? Trends in Parasitology 19, 447.Google Scholar
Tarleton, RL (2015) CD8 T cells in Trypanosoma cruzi infection. Seminars in Immunopathology 37, 233238.Google Scholar
Tarleton, RL and Zhang, L (1999) Chagas disease etiology: autoimmunity or parasite persistence? Parasitology Today 15, 9499.Google Scholar
Teixeira, RL, Hecht, MM, Guimaro, MC, Sousa, AO and Nitz, N (2011) Pathogenesis of Chagas’ disease: parasite persistence and autoimmunity. Clinical Microbiology Reviews 24, 592630.Google Scholar
Telleria, J, Biron, D, Brizard, JP, Demettre, E, Séveno, M, Barnabé, C, Ayala, FJ and Tibayrenc, M (2010) Phylogenetic character mapping of proteomic diversity shows high correlation with subspecific phylogenetic diversity in Trypanosoma cruzi. Proceedings of the National Academy of Sciences of the United States of America 107, 20411–6.Google Scholar
Vago, AR, Andrade, LO, Leite, AA, d'Avila-Reis, D, Macedo, AM, Adad, SJ, Tostes, SJ, Moreira, MC, Filho, GB and Pena, SD (2000) Genetic characterization of Trypanosoma cruzi directly from tissues of patients with chronic chagas disease: differential distribution of genetic types into diverse organs. American Journal of Pathology 156, 18051809.Google Scholar
Valadares, HM, Pimenta, JR, de Freitas, JM, Duffy, T, Bartholomeu, DC, Oliveira, RP, Chiari, E, Moreira, MC, Filho, GB, Schijman, AG, Franco, GR, Machado, CR, Pena, SD and Macedo, AM (2008) Genetic profiling of Trypanosoma cruzi directly in infected tissues using nested PCR of polymorphic microsatellites. International Journal for Parasitology 38, 839850.Google Scholar
Venegas, J, Coñoepan, W, Pichuantes, S, Miranda, S, Apt, W, Arribada, A, Zulantay, I, Coronado, X, Rodriguez, J, Reyes, E, Solari, A and Sanchez, G (2009) Differential distribution of Trypanosoma cruzi clones in human chronic chagasic cardiopathic and non-cardiopathic individuals. Acta Tropica 109, 187193.Google Scholar
Vicco, MH, Ferini, F, Rodeles, L, Cardona, P, Bontempi, I, Lioi, S, Beloscar, J, Nara, T, Marcipar, I and Bottasso, OA (2013) Assessment of cross-reactive host-pathogen antibodies in patients with different stages of chronic chagas disease. Revista Espanola de Cardiologia 66, 791796.Google Scholar
Vitelli-Avellar, DM, Sathler-Avelar, R, Mattoso-Barbosa, AM, Gouin, N, Perdigao-de-Oliveira, M, Valério-dos-Reis, L, Peres, CR, Elói-Santos, SM, Souza, GM, Rodrigues-do-Amaral, L, Teixeira-Carvahlo, A, Martins-Fihlo, OA, Dick, JEJ, Hubbard, GB, Vandenberg, JF and VandeBerg, JL (2017) Cynomolgus macaques naturally infected with Trypanosoma cruzi-I exhibit an overall mixed pro-inflammatory/modulated cytokine signature characteristic of human Chagas disease. Neglected Tropical Diseases. 11, e0005233.Google Scholar
Westenberger, SJ, Campbell, DA, Sturm, NR and Barnabé, C (2005) Two hybridization events define the population structure of Trypanosoma cruzi. Genetics 171, 527543.Google Scholar
WHO (2017) Weekly epidemiological record. World Health Organization, Relevé Épidémiologique Hebdomadaire, 92, 357368.Google Scholar
Wong, VK, Baker, S, Pickard, DJ, Parkhill, J, Page, AJ, Feasey, NA, Kingsley, RA, Thomson, NR, Keane, JA, Weill, FX, Edwards, DJ, Hawkey, J, Harris, SR, Mather, AE, Cain, AK, Hadfield, J, Hart, PJ, Thieu, NT, Klemm, EJ, Glinos, DA, Breiman, RJ, Watson, CH, Kariuki, S, Gordon, MA, Heyderman, RS, Okoro, C, Jacobs, J, Lunguya, O, Edmunds, WJ, Msefula, C, Chabalgoity, JA, Kama, M, Jenkins, K, Dutta, S, Marks, F, Campos, J, Thompson, C, Obaro, S, MacLennan, CA, Dolecek, C, Keddy, KH, Smith, AM, Parry, CM, Karkey, A, Mulholland, EK, Campbell, JI, Dongol, S, Basnyat, B, Dufour, M, Bandaranayake, D, Naseri, TT, Singh, SP, Hatta, M, Newton, P, Onsare, RS, Isaia, L, Dance, D, Davong, V, Thwaites, G, Wijedoru, L, Crump, JA, De Pinna, E, Nair, S, Nilles, EJ, Thanh, DP, Turner, P, Soeng, S, Valcanis, M, Powling, J, Dimovski, K, Hogg, G, Farrar, J, Holt, KE and Dougan, G (2015) Phylogeographical analysis of the dominant multidrug-resistant H58 clade of Salmonella Typhi identifies inter- and intracontinental transmission events. Nature Genetics 47, 632639.Google Scholar
Yeo, M, Acosta, N, Llewellyn, M, Sánchez, H, Adamson, S, Miles, GA, López, E, González, N, Patterson, JS, Gaunt, MW, de Arias, AR and Miles, MA (2005) Origins of Chagas disease: Didelphis species are natural hosts of Trypanosoma cruzi I and armadillos hosts of Trypanosoma cruzi II, including hybrids. International Journal for Parasitology 35, 225233.Google Scholar
Yeo, M, Mauricio, IL, Messenger, LA, Lewis, MD, Llewellyn, MS, Acosta, N, Bhattacharyya, T, Diosque, P, Carrasco, HJ and Miles, MA (2011) Multilocus sequence typing (MLST) for lineage assignment and high resolution diversity studies in Trypanosoma cruzi. PLoS Neglected Tropical Diseases 5, 113.Google Scholar
Yoshida, N (2006) Molecular basis of mammalian cell invasion by Trypanosoma cruzi. Anais da Academia Brasileira de Ciências 78, 87111.Google Scholar
Yoshida, N, Tyler, KM and Llewellyn, MS (2011) Invasion mechanisms among emerging food-borne protozoan parasites. Trends in Parasitology 27, 459466.Google Scholar
Zafra, G, Mantilla, JC, Jacome, J, Macedo, AM and Gonzalez, CI (2011) Direct analysis of genetic variability in Trypanosoma cruzi populations from tissues of Colombian chagasic patients. Human Pathology 42, 11591168.Google Scholar
Zingales, B (2017) Trypanosoma cruzi genetic diversity: something new for something known about Chagas disease manifestations, serodiagnosis and drug sensitivity. Acta Tropica. S0001-706X, 3042630426.Google Scholar
Zingales, B, Andrade, SG, Briones, MR, Campbell, DA, Chiari, E, Fernandes, O, Guhl, F, Lages-Silva, E, Macedo, AM, Machado, CR, Miles, MA, Romanha, AJ, Sturm, NR, Tibayrenc, M and Schijman, AG (2009) A new consensus for Trypanosoma cruzi intraspecific nomenclature: second revision meeting recommends TcI to TcVI. Memórias do Instituto Oswaldo Cruz 7, 10511054.Google Scholar
Zingales, B, Miles, MA, Campbell, DA, Tibayrenc, M, Macedo, AM, Teixeira, MM, Schijman, AG, Llewellyn, MS, Lages-Silva, E, Machado, CR, Andrade, SG and Sturm, NR (2012) The revised Trypanosoma cruzi subspecific nomenclature: rationale, epidemiological relevance and research applications. Infection, Genetics and Evolution 2, 240253.Google Scholar
Zumaya-Estrada, FA, Messenger, LA, Lopez-Ordonez, T, Lewis, MD, Flores-Lopez, CA, Martínez-Ibarra, AJ, Pennington, PM, Cordon-Rosales, C, Carrasco, HV, Segovia, M, Miles, MA and Llewellyn, MS (2012) North American import? Charting the origins of an enigmatic Trypanosoma cruzi domestic genotype. Parasites & Vectors 5, 226234.Google Scholar
Figure 0

Fig. 1. Algorithm for DTU discrimination of T. cruzi.

Figure 1

Fig. 2. Cytokine profile during T. cruzi infection. The acute phase shows the immune system response and the chronic phase shows the activation of cytokines.

Figure 2

Fig. 3. Virulence factors and immunological response elicited by T. cruzi infection.

Figure 3

Table 1. Association between the T. cruzi DTUs and the related clinical form and immune response

Figure 4

Fig. 4. Overview of the geographical distribution of T. cruzi DTUs associated with CCC across the American continent.