1
|
Bolnick DI, Arruda S, Polania C, Simonse L, Padhiar A, Rodgers ML, Roth-Monzón AJ. The Dominance of Coinfecting Parasites' Indirect Genetic Effects on Host Traits. Am Nat 2024; 204:482-500. [PMID: 39486034 DOI: 10.1086/732256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
AbstractIndirect genetic effects (IGEs) exist when there is heritable variation in one organism's ability to alter a second organism's traits. For example, parasites have antigens that can induce a host immune response, as well as disparate strategies to evade or suppress host immunity; among-parasite genetic variation in these antigens generates among-host variation in immune traits. Here, we experimentally show that the cestode parasite Schistocephalus solidus exerts an IGE on an immune trait (peritoneal fibrosis) in its threespine stickleback host: stickleback developed strong fibrosis after exposure to some parasite genotypes but not others. A complication arises during coinfection, when two or more parasite genotypes may impose conflicting IGEs on the same host trait. What parasite-controlled trait will the host express? Will the host trait reflect the more immune-stimulatory parasite genotype or the more immune-evasive genotype? These alternatives can be quantified by estimating the dominance coefficient, as if a coinfected host were a heterozygote. We experimentally estimated the dominance of S. solidus IGEs by coinjecting antigens from different parasite genotypes. Contrary to our a priori hypotheses, coinjected antigens induced an overdominant effect, stronger than either parasite's antigens alone. We present a mathematical model showing that the value of this IGE dominance is biologically important, affecting the evolutionary dynamics of parasites in a density- and frequency-dependent manner. The model indicates that overdominance would be detrimental to immigrants when resident prevalence is high. This combination of experimental data and modeling provides an example of a parasite IGE on host traits and the evolutionary significance of IGE dominance.
Collapse
|
2
|
Silvestrini MMA, Alessio GD, Frias BED, Sales Júnior PA, Araújo MSS, Silvestrini CMA, Brito Alvim de Melo GE, Martins-Filho OA, Teixeira-Carvalho A, Martins HR. New insights into Trypanosoma cruzi genetic diversity, and its influence on parasite biology and clinical outcomes. Front Immunol 2024; 15:1342431. [PMID: 38655255 PMCID: PMC11035809 DOI: 10.3389/fimmu.2024.1342431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 04/26/2024] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, remains a serious public health problem worldwide. The parasite was subdivided into six distinct genetic groups, called "discrete typing units" (DTUs), from TcI to TcVI. Several studies have indicated that the heterogeneity of T. cruzi species directly affects the diversity of clinical manifestations of Chagas disease, control, diagnosis performance, and susceptibility to treatment. Thus, this review aims to describe how T. cruzi genetic diversity influences the biology of the parasite and/or clinical parameters in humans. Regarding the geographic dispersion of T. cruzi, evident differences were observed in the distribution of DTUs in distinct areas. For example, TcII is the main DTU detected in Brazilian patients from the central and southeastern regions, where there are also registers of TcVI as a secondary T. cruzi DTU. An important aspect observed in previous studies is that the genetic variability of T. cruzi can impact parasite infectivity, reproduction, and differentiation in the vectors. It has been proposed that T. cruzi DTU influences the host immune response and affects disease progression. Genetic aspects of the parasite play an important role in determining which host tissues will be infected, thus heavily influencing Chagas disease's pathogenesis. Several teams have investigated the correlation between T. cruzi DTU and the reactivation of Chagas disease. In agreement with these data, it is reasonable to suppose that the immunological condition of the patient, whether or not associated with the reactivation of the T. cruzi infection and the parasite strain, may have an important role in the pathogenesis of Chagas disease. In this context, understanding the genetics of T. cruzi and its biological and clinical implications will provide new knowledge that may contribute to additional strategies in the diagnosis and clinical outcome follow-up of patients with Chagas disease, in addition to the reactivation of immunocompromised patients infected with T. cruzi.
Collapse
Affiliation(s)
| | - Glaucia Diniz Alessio
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Bruna Estefânia Diniz Frias
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Policarpo Ademar Sales Júnior
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Márcio Sobreira Silva Araújo
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Olindo Assis Martins-Filho
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Andréa Teixeira-Carvalho
- Integrated Biomarker Research Group, René Rachou Institute, Fiocruz Minas, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
| | - Helen Rodrigues Martins
- Department of Pharmacy, Federal University of the Jequitinhonha and Mucuri Valleys, Diamantina, Minas Gerais, Brazil
| |
Collapse
|
3
|
Dumonteil E, Desale H, Tu W, Hernandez-Cuevas N, Shroyer M, Goff K, Marx PA, Herrera C. Intra-host Trypanosoma cruzi strain dynamics shape disease progression: the missing link in Chagas disease pathogenesis. Microbiol Spectr 2023; 11:e0423622. [PMID: 37668388 PMCID: PMC10581044 DOI: 10.1128/spectrum.04236-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/10/2023] [Indexed: 09/06/2023] Open
Abstract
Chronic Chagasic cardiomyopathy develops years after infection in 20-40% of patients, but disease progression is poorly understood. Here, we assessed Trypanosoma cruzi parasite dynamics and pathogenesis over a 2.5-year period in naturally infected rhesus macaques. Individuals with better control of parasitemia were infected with a greater diversity of parasite strains compared to those with increasing parasitemia over time. Also, the in vivo parasite multiplication rate decreased with increasing parasite diversity, suggesting competition among strains or a stronger immune response in multiple infections. Significant differences in electrocardiographic (ECG) profiles were observed in Chagasic macaques compared to uninfected controls, suggesting early conduction defects, and changes in ECG patterns over time were observed only in macaques with increasing parasitemia and lower parasite diversity. Disease progression was also associated with plasma fibronectin degradation, which may serve as a biomarker. These data provide a novel framework for the understanding of Chagas disease pathogenesis, with parasite diversity shaping disease progression.IMPORTANCEChagas disease progression remains poorly understood, and patients at increased risk of developing severe cardiac disease cannot be distinguished from those who may remain asymptomatic. Monitoring of Trypanosoma cruzi strain dynamics and pathogenesis over 2-3 years in naturally infected macaques shows that increasing parasite diversity in hosts is detrimental to parasite multiplication and Chagasic cardiomyopathy disease progression. This provides a novel framework for the understanding of Chagas disease pathogenesis.
Collapse
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Hans Desale
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Weihong Tu
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Nora Hernandez-Cuevas
- Laboratorio de Parasitologia, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Merida, Yucatan, Mexico
| | - Monica Shroyer
- Division of Veterinary Medicine, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Preston A. Marx
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Claudia Herrera
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
4
|
Koh CC, Neves EGA, de Souza-Silva TG, Carvalho AC, Pinto CHR, Sobreira Galdino A, Gollob KJ, Dutra WO. Cytokine Networks as Targets for Preventing and Controlling Chagas Heart Disease. Pathogens 2023; 12:171. [PMID: 36839443 PMCID: PMC9966322 DOI: 10.3390/pathogens12020171] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Chagas disease, a neglected disease caused by the protozoan Trypanosoma cruzi, is endemic in 21 Latin American countries, affecting 6-8 million people. Increasing numbers of Chagas disease cases have also been reported in non-endemic countries due to migration, contamination via blood transfusions or organ transplantation, characterizing Chagas as an emerging disease in such regions. While most individuals in the chronic phase of Chagas disease remain in an asymptomatic clinical form named indeterminate, approximately 30% of the patients develop a cardiomyopathy that is amongst the deadliest cardiopathies known. The clinical distinctions between the indeterminate and the cardiac clinical forms are associated with different immune responses mediated by innate and adaptive cells. In this review, we present a collection of studies focusing on the human disease, discussing several aspects that demonstrate the association between chemokines, cytokines, and cytotoxic molecules with the distinct clinical outcomes of human infection with Trypanosoma cruzi. In addition, we discuss the role of gene polymorphisms in the transcriptional control of these immunoregulatory molecules. Finally, we discuss the potential application of cytokine expression and gene polymorphisms as markers of susceptibility to developing the severe form of Chagas disease, and as targets for disease control.
Collapse
Affiliation(s)
- Carolina Cattoni Koh
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Eula G. A. Neves
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Thaiany Goulart de Souza-Silva
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Ana Carolina Carvalho
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Cecília Horta Ramalho Pinto
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Alexsandro Sobreira Galdino
- Laboratório de Biotecnologia de Microrganismos, Universidade Federal de São João Del-Rei (UFSJ), Campus Centro Oeste, Divinópolis 35501-296, MG, Brazil
| | - Kenneth J. Gollob
- Hospital Israelita Albert Einstein, São Paulo 05652-900, SP, Brazil
- Instituto Nacional de Ciências e Tecnologia em Doenças Tropicais, INCT-DT, Salvador 40110-160, BA, Brazil
| | - Walderez Ornelas Dutra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
- Instituto Nacional de Ciências e Tecnologia em Doenças Tropicais, INCT-DT, Salvador 40110-160, BA, Brazil
| |
Collapse
|
5
|
SOCS2 expression in hematopoietic and non-hematopoietic cells during Trypanosoma cruzi infection: Correlation with immune response and cardiac dysfunction. Clin Immunol 2021; 234:108913. [PMID: 34954347 DOI: 10.1016/j.clim.2021.108913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/16/2021] [Accepted: 12/18/2021] [Indexed: 11/23/2022]
Abstract
Chagas disease has a complex pathogenesis wherein the host immune response is essential for controlling its development. Suppressor of cytokine signaling(SOCS)2 is a crucial protein that regulates cytokine production. In this study, SOCS2 deficiency resulted in an initial imbalance of IL12- and IL-10-producing neutrophils and dendritic cells (DCs), which caused a long-lasting impact reducing inflammatory neutrophils and DCs, and tolerogenic DCs at the peak of acute disease. A reduced number of inflammatory and pro-resolving macrophages, and IL17A-producing CD4+ T cells, and increased lymphocyte apoptosis was found in SOCS2-deficient mice. Electrocardiogram analysis of chimeric mice showed that WT mice that received SOCS2 KO bone marrow transplantation presented increased heart dysfunction. Taken together, the results demonstrated that SOCS2 is a crucial regulator of the immune response during Trypanosoma cruzi infection, and suggest that a SOCS2 genetic polymorphism, or failure of its expression, may increase the susceptibility of cardiomyopathy development in Chagasic patients.
Collapse
|
6
|
Chagas disease: Immunology of the disease at a glance. Cytokine Growth Factor Rev 2021; 62:15-22. [PMID: 34696979 DOI: 10.1016/j.cytogfr.2021.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Chagas disease is an important neglected disease that affects 6-7 million people worldwide. The disease has two phases: acute and chronic, in which there are different clinical symptoms. Controlling the infection depends on innate and acquired immune responses, which are activated during the initial infection and are critical for host survival. Furthermore, the immune system plays an important role in the therapeutic success. Here we summarize the importance of the immune system cytokines in the pathology outcome, as well as in the treatment.
Collapse
|
7
|
da Silveira-Lemos D, Alessio GD, Batista MA, de Azevedo PO, Reis-Cunha JL, Mendes TADO, Lourdes RDA, de Lana M, Fujiwara RT, Filho OAM, Bartholomeu DC. Phenotypic, functional and serological aspects of genotypic-specific immune response of experimental T. cruzi infection. Acta Trop 2021; 222:106021. [PMID: 34161815 DOI: 10.1016/j.actatropica.2021.106021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
The complexity and multifactorial characteristics of Chagas disease pathogenesis hampers the establishment of appropriate experimental/epidemiological sets, and therefore, still represents one of the most challenging fields for novel insights and discovery. In this context, we used a set of attributes including phenotypic, functional and serological markers of immune response as candidates to decode the genotype-specific immune response of experimental T. cruzi infection. In this investigation, we have characterized in C57BL/6 J mice, the early (parasitemia peak) and late (post-parasitemia peak) aspects of the immune response elicited by T. cruzi strains representative of TcI, TcII or TcVI. The results demonstrated earlier parasitemia peak for TcII/Y strain followed by TcVI/CL-Brener and TcI/Colombiana strains. A panoramic overview of phenotypic and functional features of the TCD4+, TCD8+ and B-cells from splenocytes demonstrated that mice infected with TcI/Colombiana strain exhibited at early stages of infection low levels of most cytokine+ cells with a slight increase at late stages of infection. Conversely, mice infected with TcII/Y strain presented an early massive increase of cytokine+ cells, which decreases at late stages. The TcVI/CL-Brener strain showed an intermediate profile at early stages of infection with a slight increase later on at post-peak of parasitemia. The panoramic analysis of immunological connectivity demonstrated that early after infection, the TcI/Colombiana strain trigger immunological network characterized by a small number of connectivity, selectively amongst cytokines that further shade towards the late stages of infection. In contrast, the TcII/Y strain elicited in more imbricate networks early after infection, comprising a robust number of interactions between pro-inflammatory mediators, regulatory cytokines and activation markers that also decrease at late infection. On the other hand, the infection with TcVI/CL-Brener strain demonstrated an intermediate profile with connectivity axes more stable at early and late stages of infection. The analysis of IgG2a reactivity to AMA, TRYPO and EPI antigens revealed that at early stages of infection, the genotype-specific reactivity to AMA, TRYPO and EPI to distinguish was higher for TcI/Colombiana as compared to TcII/Y and TcVI/CL while, at late stages of infection, higher reactivity to AMA was observed in mice infected with TcVI/CL and TcII/Y strains. The novel systems biology approaches and the use of a flow cytometry platform demonstrated that distinct T. cruzi genotypes influenced in the phenotypic and functional features of the host immune response and the genotype-specific serological reactivity during early and late stages of experimental T. cruzi infection.
Collapse
Affiliation(s)
- Denise da Silveira-Lemos
- Laboratório de Imunologia e Genômica de Parasitos - Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Minas Gerais, Brasil; Grupo Integrado de Pesquisas em Biomarcadores - Instituto René Rachou/Fiocruz-MINAS, Minas Gerais, Brasil.
| | - Glaucia Diniz Alessio
- Grupo Integrado de Pesquisas em Biomarcadores - Instituto René Rachou/Fiocruz-MINAS, Minas Gerais, Brasil; Campus Centro-Oeste Dona Lindu, Universidade Federal de São João Del-Rei, Divinópolis, Minas Gerais, Brasil.
| | - Maurício Azevedo Batista
- Laboratório de Imunologia e Genômica de Parasitos - Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Patrick Orestes de Azevedo
- Grupo Integrado de Pesquisas em Biomarcadores - Instituto René Rachou/Fiocruz-MINAS, Minas Gerais, Brasil
| | - João Luís Reis-Cunha
- Laboratório de Imunologia e Genômica de Parasitos - Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Tiago Antônio de Oliveira Mendes
- Laboratório de Imunologia e Genômica de Parasitos - Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Rodrigo de Almeida Lourdes
- Laboratório de Imunologia e Genômica de Parasitos - Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Marta de Lana
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto, Minas Gerais, Brasil
| | - Ricardo Toshio Fujiwara
- Laboratório de Imunologia e Genômica de Parasitos - Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Olindo Assis Martins Filho
- Grupo Integrado de Pesquisas em Biomarcadores - Instituto René Rachou/Fiocruz-MINAS, Minas Gerais, Brasil
| | - Daniella Castanheira Bartholomeu
- Laboratório de Imunologia e Genômica de Parasitos - Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| |
Collapse
|
8
|
de Castro TBR, Canesso MCC, Boroni M, Chame DF, Souza DDL, de Toledo NE, Tahara EB, Pena SD, Machado CR, Chiari E, Macedo A, Franco GR. Differential Modulation of Mouse Heart Gene Expression by Infection With Two Trypanosoma cruzi Strains: A Transcriptome Analysis. Front Genet 2020; 11:1031. [PMID: 33088283 PMCID: PMC7495023 DOI: 10.3389/fgene.2020.01031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/11/2020] [Indexed: 12/17/2022] Open
Abstract
The protozoan Trypanosoma cruzi (T. cruzi) is a well-adapted parasite to mammalian hosts and the pathogen of Chagas disease in humans. As both host and T. cruzi are highly genetically diverse, many variables come into play during infection, making disease outcomes difficult to predict. One important challenge in the field of Chagas disease research is determining the main factors leading to parasite establishment in the chronic stage in some organs, mainly the heart and/or digestive system. Our group previously showed that distinct strains of T. cruzi (JG and Col1.7G2) acquired differential tissue distribution in the chronic stage in dually infected BALB/c mice. To investigate changes in the host triggered by the two distinct T. cruzi strains, we assessed the gene expression profiles of BALB/c mouse hearts infected with either JG, Col1.7G2 or an equivalent mixture of both parasites during the initial phase of infection. This study demonstrates the clear differences in modulation of host gene expression by both parasites. Col1.7G2 strongly activated Th1-polarized immune signature genes, whereas JG caused only minor activation of the host immune response. Moreover, JG strongly reduced the expression of genes encoding ribosomal proteins and mitochondrial proteins related to the electron transport chain. Interestingly, the evaluation of gene expression in mice inoculated with a mixture of the parasites produced expression profiles with both up- and downregulated genes, indicating the coexistence of both parasite strains in the heart during the acute phase. This study suggests that different strains of T. cruzi may be distinguished by their efficiency in activating the immune system, modulating host energy metabolism and reactive oxygen species production and decreasing protein synthesis during early infection, which may be crucial for parasite persistence in specific organs.
Collapse
Affiliation(s)
| | | | - Mariana Boroni
- Laboratório de Bioinformática e Biologia Computacional, Centro de Pesquisas, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Daniela Ferreira Chame
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Daniela de Laet Souza
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Nayara Evelin de Toledo
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Eric Birelli Tahara
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Sergio Danilo Pena
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Carlos Renato Machado
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Egler Chiari
- Departamento de Parasitologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Andrea Macedo
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| | - Gloria Regina Franco
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, UFMG, Belo Horizonte, Brazil
| |
Collapse
|
9
|
Ayala EV, Rodrigues da Cunha G, Azevedo MA, Calderon M, Jimenez J, Venuto AP, Gazzinelli R, Lavalle RJY, Riva AGV, Hincapie R, Finn MG, Marques AF. C57BL/6 α-1,3-Galactosyltransferase Knockout Mouse as an Animal Model for Experimental Chagas Disease. ACS Infect Dis 2020; 6:1807-1815. [PMID: 32374586 DOI: 10.1021/acsinfecdis.0c00061] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The leading animal model of experimental Chagas disease, the mouse, plays a significant role in studies for vaccine development, diagnosis, and human therapies. Humans, along with Old World primates, alone among mammals, cannot make the terminal carbohydrate linkage of the α-Gal trisaccharide. It has been established that the anti-α-Gal immune response is likely to be a critical factor for protection against Trypanosoma cruzi (T. cruzi) infection in humans. However, the mice customarily employed for the study of T. cruzi infection naturally express the α-Gal epitope and therefore do not produce anti-α-Gal antibodies. Here, we used the C57BL/6 α-1,3-galactosyltransferase knockout (α-GalT-KO) mouse, which does not express the α-Gal epitope as a model for experimental Chagas disease. We found the anti-α-Gal IgG antibody response to an increase in α-GalT-KO mice infected with Arequipa and Colombiana strains of T. cruzi, leading to fewer parasite nests, lower parasitemia, and an increase of INF-γ, TNF-α, and IL-12 cytokines in the heart of α-GalT-KO mice compared with α-GalT-WT mice on days 60 and 120 postinfection. We therefore agree that the C57BL/6 α-GalT-KO mouse represents a useful model for initial testing of therapeutic and immunological approaches against different strains of T. cruzi.
Collapse
Affiliation(s)
- Edward Valencia Ayala
- Laboratório de Imunologia e Genômica de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
- Instituto de Investigación, Centro de Investigación en Inmunología e Infectología, Facultad de Medicina Humana, Universidad de San Martin de Porres, Lima 15000, Perú
| | - Gisele Rodrigues da Cunha
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Maira Araujo Azevedo
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Maritza Calderon
- Laboratorio de Investigación en Enfermedades Infecciosas and Laboratorio de Biología Molecular, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15000, Perú
| | - Juan Jimenez
- Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima 15000, Perú
| | - Ana Paula Venuto
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| | - Ricardo Gazzinelli
- Instituto de Pesquisa Rene Rachou, Fundacao Oswaldo Cruz, Belo Horizonte, Minas Gerais 30190-009, Brazil
- Plataforma de Medicina Translacional, Fundacao Oswaldo Cruz, Belo Horizonte, Minas Gerais 30190-009, Brazil
| | - Raúl Jesus Ynocente Lavalle
- Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima 15000, Perú
| | - Angela Giovana Vidal Riva
- Instituto de Investigación, Centro de Investigación en Inmunología e Infectología, Facultad de Medicina Humana, Universidad de San Martin de Porres, Lima 15000, Perú
- Laboratorio de Investigación en Enfermedades Infecciosas and Laboratorio de Biología Molecular, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15000, Perú
| | - Robert Hincapie
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332 United States
| | - M. G. Finn
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332 United States
| | - Alexandre F. Marques
- Laboratório de Imuno-Proteômica e Biologia de Parasitos, Departamento de Parasitologia, Instituto de Ciências Biológicas/ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270901, Brazil
| |
Collapse
|
10
|
Rojo G, Pèlissier F, Sandoval-Rodriguez A, Bacigalupo A, García V, Pinto R, Ortiz S, Botto-Mahan C, Cattan PE, Solari A. Organs infected with Trypanosoma cruzi and DTU identification in the naturally infected rodent Octodon degus. Exp Parasitol 2020; 215:107931. [PMID: 32464222 DOI: 10.1016/j.exppara.2020.107931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 11/19/2022]
Abstract
Chagas disease is a public health problem in America. Its parasite, Trypanosoma cruzi, presents different discrete typing units (DTUs), colonizes organs of mammalian hosts in chronic infections, and presents tropism for particular organs in experimental infections. We evaluated T. cruzi tropism towards organs on the naturally infected rodent Octodon degus, identifying the parasites' DTUs, by means of conventional PCR and hybridization. Almost all the analyzed organs presented T. cruzi. More than 42% of the tested oesophagus, skin, skeletal muscle, brain and intestine showed T. cruzi DNA. Other nine types of organs were infected in over 15%. These results suggest that there is some tropism by T. cruzi in chronically infected O. degus. DTU TcV was present in 92.5% of infected organs with identified DTUs; this DTU is frequently reported in human infections in the Southern Cone of South America. Few organs showed mixed DTU infections. This is one of the few reports on the outcome of chronic natural T. cruzi-infection in wild mammal hosts exposed to naturally infected vectors.
Collapse
Affiliation(s)
- Gemma Rojo
- Instituto de Ciencias Agroalimentarias, Animales y Ambientales (ICA3), Universidad de O'Higgins, Rancagua, Chile; Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Francisca Pèlissier
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile.
| | - Alejandra Sandoval-Rodriguez
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile.
| | - Antonella Bacigalupo
- Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile; Institute of Biodiversity, Animal Health & Comparative Medicine, University of Glasgow, Glasgow, United Kingdom.
| | - Vanessa García
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile.
| | - Raquel Pinto
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile.
| | - Sylvia Ortiz
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| | - Carezza Botto-Mahan
- Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.
| | - Pedro E Cattan
- Departamento de Ciencias Biológicas Animales, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile.
| | - Aldo Solari
- Programa de Biología Celular y Molecular, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
11
|
Maurya R, Bhattacharya P, Dey R, Nakhasi HL. Leptin Functions in Infectious Diseases. Front Immunol 2018; 9:2741. [PMID: 30534129 PMCID: PMC6275238 DOI: 10.3389/fimmu.2018.02741] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 11/07/2018] [Indexed: 01/26/2023] Open
Abstract
Leptin, a pleiotropic protein has long been recognized to play an important role in the regulation of energy homeostasis, metabolism, neuroendocrine function, and other physiological functions through its effects on the central nervous system (CNS) and peripheral tissues. Leptin is secreted by adipose tissue and encoded by the obese (ob) gene. Leptin acts as a central mediator which regulates immunity as well as nutrition. Importantly, leptin can modulate both innate and adaptive immune responses. Leptin deficiency/resistance is associated with dysregulation of cytokine production, increased susceptibility toward infectious diseases, autoimmune disorders, malnutrition and inflammatory responses. Malnutrition induces a state of immunodeficiency and an inclination to death from communicable diseases. Infectious diseases are the disease of poor who invariably suffer from malnutrition that could result from reduced serum leptin levels. Thus, leptin has been placed at the center of many interrelated functions in various pathogenic conditions, such as bacterial, viruses and parasitic infections. We review herein, the recent advances on the role of leptin in malnutrition in pathogenesis of infectious diseases with a particular emphasis on parasitic diseases such as Leishmaniasis, Trypanosomiasis, Amoebiasis, and Malaria.
Collapse
Affiliation(s)
- Radheshyam Maurya
- Department of Animal Biology, School of Life Science, University of Hyderabad, Hyderabad, India
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Hira L. Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
12
|
Who benefits from cellular immune response during the Chagas disease? Biosystems 2018; 171:66-73. [PMID: 30055256 DOI: 10.1016/j.biosystems.2018.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 11/22/2022]
Abstract
We extend our previous model for the dynamical interaction between a mammal's immune response and the Trypanosoma cruzi parasite during the acute phase of Chagas disease. The model here considers both humoral and cellular responses and the different stages of T. cruzi (intracellular and extracellular phases) inside the mammal host. We analyze the dynamical time evolution of the populations obtaining phase diagrams of the model results. The steady-state solution of the system yields two outcomes associated to Healing and Chronic stationary cases, the death case obtained when just the humoral immune response alone was considered is not being present. This result implies that, surprisingly, although the immune cellular response is obviously beneficial for the host, it is also evolutionary advantageous for the parasite, as it helps to preserve the host alive and, after transmission to a healthy host, perpetuate the disease. Of course, if the cell damage by the parasite's intracellular stage is high, it may cause the host death. This possibility is accounted in the model by introducing a death criterion related to cell destruction. We present a new phase diagram, that restores the host death case and generates a phase diagram similar to the one arising from the original model.
Collapse
|
13
|
Strauss M, Velázquez López DA, Moya DM, Bazán PC, Báez AL, Rivarola HW, Paglini-Oliva PA, Lo Presti MS. Differential tissue distribution of Trypanosoma cruzi during acute experimental infection: Further evidence using natural isolates. Mol Biochem Parasitol 2018; 222:29-33. [PMID: 29709547 DOI: 10.1016/j.molbiopara.2018.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 10/17/2022]
Abstract
In the present work, we evaluated the effect of mixed Trypanosoma cruzi infections, studying the biological distribution of the different parasites in blood, heart and skeletal muscle during the acute phase. Albino Swiss mice were infected with different parasite strain/isolates or with a combination of them. The parasites in the different tissues were typified through specific PCR, population variability was analyzed through RFLP studies and parasitological and histopathological parameters were evaluated. We found a predominance of TcII and TcVI in all tissues samples respect to TcV and different parasite populations were found in circulation and in the tissues from the same host. These results verify the distribution of parasites in host tissues from early stages of infection and show biological interactions among different genotypes and populations of T. cruzi.
Collapse
Affiliation(s)
- Mariana Strauss
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina
| | - Daniela A Velázquez López
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina
| | - Diego M Moya
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina
| | - P Carolina Bazán
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina
| | - Alejandra L Báez
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina
| | - H Walter Rivarola
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina
| | - Patricia A Paglini-Oliva
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina
| | - M Silvina Lo Presti
- Instituto de Investigaciones en Ciencias de la Salud (INICSA) UNC - CONICET, Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, Cátedra de Física Biomédica, Facultad de Ciencias Médicas, Córdoba, Argentina.
| |
Collapse
|
14
|
Dario MA, Moratelli R, Schwabl P, Jansen AM, Llewellyn MS. Small subunit ribosomal metabarcoding reveals extraordinary trypanosomatid diversity in Brazilian bats. PLoS Negl Trop Dis 2017; 11:e0005790. [PMID: 28727769 PMCID: PMC5544246 DOI: 10.1371/journal.pntd.0005790] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/04/2017] [Accepted: 07/10/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Bats are a highly successful, globally dispersed order of mammals that occupy a wide array of ecological niches. They are also intensely parasitized and implicated in multiple viral, bacterial and parasitic zoonoses. Trypanosomes are thought to be especially abundant and diverse in bats. In this study, we used 18S ribosomal RNA metabarcoding to probe bat trypanosome diversity in unprecedented detail. METHODOLOGY/PRINCIPAL FINDINGS Total DNA was extracted from the blood of 90 bat individuals (17 species) captured along Atlantic Forest fragments of Espírito Santo state, southeast Brazil. 18S ribosomal RNA was amplified by standard and/or nested PCR, then deep sequenced to recover and identify Operational Taxonomic Units (OTUs) for phylogenetic analysis. Blood samples from 34 bat individuals (13 species) tested positive for infection by 18S rRNA amplification. Amplicon sequences clustered to 14 OTUs, of which five were identified as Trypanosoma cruzi I, T. cruzi III/V, Trypanosoma cruzi marinkellei, Trypanosoma rangeli, and Trypanosoma dionisii, and seven were identified as novel genotypes monophyletic to basal T. cruzi clade types of the New World. Another OTU was identified as a trypanosome like those found in reptiles. Surprisingly, the remaining OTU was identified as Bodo saltans-closest non-parasitic relative of the trypanosomatid order. While three blood samples featured just one OTU (T. dionisii), all others resolved as mixed infections of up to eight OTUs. CONCLUSIONS/SIGNIFICANCE This study demonstrates the utility of next-generation barcoding methods to screen parasite diversity in mammalian reservoir hosts. We exposed high rates of local bat parasitism by multiple trypanosome species, some known to cause fatal human disease, others non-pathogenic, novel or yet little understood. Our results highlight bats as a long-standing nexus among host-parasite interactions of multiple niches, sustained in part by opportunistic and incidental infections of consequence to evolutionary theory as much as to public health.
Collapse
Affiliation(s)
- Maria Augusta Dario
- Laboratório de Biologia de Tripanosomatídeos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Moratelli
- Fiocruz Mata Atlântica, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Philipp Schwabl
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Ana Maria Jansen
- Laboratório de Biologia de Tripanosomatídeos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martin S. Llewellyn
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
15
|
Cooper C, Clode PL, Peacock C, Thompson RCA. Host-Parasite Relationships and Life Histories of Trypanosomes in Australia. ADVANCES IN PARASITOLOGY 2016; 97:47-109. [PMID: 28325373 DOI: 10.1016/bs.apar.2016.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Trypanosomes constitute a group of flagellate protozoan parasites responsible for a number of important, yet neglected, diseases in both humans and livestock. The most significantly studied include the causative agents of African sleeping sickness (Trypanosoma brucei) and Chagas disease (Trypanosoma cruzi) in humans. Much of our knowledge about trypanosome host-parasite relationships and life histories has come from these two human pathogens. Recent investigations into the diversity and life histories of wildlife trypanosomes in Australia highlight that there exists a great degree of biological and behavioural variation within and between trypanosomes. In addition, the genetic relationships between some Australian trypanosomes show that they are unexpectedly more closely related to species outside Australia than within it. These findings have led to a growing focus on the importance of understanding parasites occurring naturally in wildlife to (1) better document parasite biodiversity, (2) determine evolutionary relationships and degree of host specificity, (3) understand host-parasite interactions and the role of parasites in the natural ecosystem and (4) identify biosecurity issues of emerging disease in both wildlife and human populations. Here we review what is known about the diversity, life histories, host-parasite interactions and evolutionary relationships of trypanosomes in Australian wildlife. In this context, we focus upon the genetic proximity of key Australian species to the pathogenic T. cruzi and discuss similarities in their biology and behaviour that present a potential risk of human disease transmission by Australian vectors and wildlife.
Collapse
Affiliation(s)
- C Cooper
- The University of Western Australia, Crawley, WA, Australia
| | - P L Clode
- The University of Western Australia, Crawley, WA, Australia
| | - C Peacock
- The University of Western Australia, Crawley, WA, Australia; Telethon Kids Institute, Subiaco, WA, Australia
| | | |
Collapse
|
16
|
Melero-Jerez C, Ortega MC, Moliné-Velázquez V, Clemente D. Myeloid derived suppressor cells in inflammatory conditions of the central nervous system. Biochim Biophys Acta Mol Basis Dis 2015; 1862:368-80. [PMID: 26527182 DOI: 10.1016/j.bbadis.2015.10.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/16/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022]
Abstract
The knowledge of the immune system elements and their relationship with other tissues, organs and systems are key approximations for the resolution of many immune-related disorders. The control of the immune response and/or its modulation from the pro-inflammatory to the anti-inflammatory response is being deeply studied in the field. In the last years, the study of myeloid-derived suppressor cells (MDSCs), a group of immature myeloid cells with a high suppressive activity on T cells has been extensively addressed in cancer. In contrast, their role in neuroimmune diseases is far from being totally understood. In this review, we will summarize data about MDSCs coming from the study of neuroinflammatory diseases in general and their potential role in multiple sclerosis, in order to introduce the putative use of this extraordinary promising cell type for future cell-based therapies. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Carolina Melero-Jerez
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda" s/n, E-45071 Toledo, Spain
| | - María Cristina Ortega
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda" s/n, E-45071 Toledo, Spain; Centro de Biología Molecular Severo Ochoa. Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Verónica Moliné-Velázquez
- Animal Experimental Unit, Scientific Instrumentation Center (CIC), Campus de la Cartuja, Universidad de Granada, Granada, Spain
| | - Diego Clemente
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca "La Peraleda" s/n, E-45071 Toledo, Spain.
| |
Collapse
|
17
|
Nogueira-Paiva NC, Vieira PMDA, Oliveri LMR, Fonseca KDS, Pound-Lana G, de Oliveira MT, de Lana M, Veloso VM, Reis AB, Tafuri WL, Carneiro CM. Host-Parasite Interactions in Chagas Disease: Genetically Unidentical Isolates of a Single Trypanosoma cruzi Strain Identified In Vitro via LSSP-PCR. PLoS One 2015; 10:e0137788. [PMID: 26359864 PMCID: PMC4567304 DOI: 10.1371/journal.pone.0137788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/21/2015] [Indexed: 11/18/2022] Open
Abstract
The present study aims at establishing whether the diversity in pathogenesis within a genetically diverse host population infected with a single polyclonal strain of Trypanosoma cruzi is due to selection of specific subpopulations within the strain. For this purpose we infected Swiss mice, a genetically diverse population, with the polyclonal strain of Trypanosoma cruzi Berenice-78 and characterized via LSSP-PCR the kinetoplast DNA of subpopulations isolated from blood samples collected from the animals at various times after inoculation (3, 6 and 12 months after inoculation). We examined the biological behavior of the isolates in acellular medium and in vitro profiles of infectivity in Vero cell medium. We compared the characteristics of the isolates with the inoculating strain and with another strain, Berenice 62, isolated from the same patient 16 years earlier. We found that one of the isolates had intermediate behavior in comparison with Berenice-78 and Berenice-62 and a significantly different genetic profile by LSSP-PCR in comparison with the inoculating strain. We hereby demonstrate that genetically distinct Trypanosoma cruzi isolates may be obtained upon experimental murine infection with a single polyclonal Trypanosoma cruzi strain.
Collapse
Affiliation(s)
- Nívia Carolina Nogueira-Paiva
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Paula Melo de Abreu Vieira
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
- Departamento de Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Larissa Maris Rezende Oliveri
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Kátia da Silva Fonseca
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Gwenaelle Pound-Lana
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Maykon Tavares de Oliveira
- Laboratório de Doença de Chagas, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto,Ouro Preto, MG, Brazil
| | - Marta de Lana
- Laboratório de Doença de Chagas, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto,Ouro Preto, MG, Brazil
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto,Ouro Preto, MG, Brazil
| | - Vanja Maria Veloso
- Departamento de Farmácia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Alexandre Barbosa Reis
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto,Ouro Preto, MG, Brazil
| | - Washington Luiz Tafuri
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
| | - Cláudia Martins Carneiro
- Laboratório de Imunopatologia, Núcleo de Pesquisas em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, MG, Brazil
- Departamento de Análises Clínicas, Escola de Farmácia, Universidade Federal de Ouro Preto,Ouro Preto, MG, Brazil
- * E-mail:
| |
Collapse
|
18
|
Peterson JK, Graham AL, Dobson AP, Chávez OT. Rhodnius prolixus Life History Outcomes Differ when Infected with Different Trypanosoma cruzi I Strains. Am J Trop Med Hyg 2015; 93:564-72. [PMID: 26078316 DOI: 10.4269/ajtmh.15-0218] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/02/2015] [Indexed: 12/20/2022] Open
Abstract
The effect of a parasite on the life history of its vector is important for understanding and predicting disease transmission. Chagas disease agent Trypanosoma cruzi is a generalist parasite that is diverse across scales from its genetic diversity to the 100s of mammal and vector species it infects. Its vertebrate hosts show quite variable responses to infection, however, to date there are no studies looking at how T. cruzi variability might result in variable outcomes in its invertebrate host. Therefore, we investigated the effect of different T. cruzi I strains on Rhodnius prolixus survival and development. We found significant variation between insects infected with different strains, with some strains having no effect, as compared with uninfected insects, and others with significantly lower survival and development. We also found that different variables had varying importance between strains, with the effect of time postinfection and the blood:weight ratio of the infective meal significantly affecting the survival of insects infected with some strains, but not others. Our results suggest that T. cruzi can be pathogenic not only to its vertebrate hosts but also to its invertebrate hosts.
Collapse
Affiliation(s)
- Jennifer K Peterson
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| | - Andrea L Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| | - Andrew P Dobson
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| | - Omar Triana Chávez
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey; Grupo BCEI, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
19
|
Ragone PG, Pérez Brandán C, Monje Rumi M, Tomasini N, Lauthier JJ, Cimino RO, Uncos A, Ramos F, Alberti D´Amato AM, Basombrío MA, Diosque P. Experimental evidence of biological interactions among different isolates of Trypanosoma cruzi from the Chaco Region. PLoS One 2015; 10:e0119866. [PMID: 25789617 PMCID: PMC4366099 DOI: 10.1371/journal.pone.0119866] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 01/16/2015] [Indexed: 01/06/2023] Open
Abstract
Many infectious diseases arise from co-infections or re-infections with more than one genotype of the same pathogen. These mixed infections could alter host fitness, the severity of symptoms, success in pathogen transmission and the epidemiology of the disease. Trypanosoma cruzi, the etiological agent of Chagas disease, exhibits a high biological variability often correlated with its genetic diversity. Here, we developed an experimental approach in order to evaluate biological interaction between three T. cruzi isolates belonging to different Discrete Typing Units (DTUs TcIII, TcV and TcVI). These isolates were obtained from a restricted geographical area in the Chaco Region. Different mixed infections involving combinations of two isolates (TcIII + TcV, TcIII + TcVI and TcV + TcVI) were studied in a mouse model. The parameters evaluated were number of parasites circulating in peripheral blood, histopathology and genetic characterization of each DTU in different tissues by DNA hybridization probes. We found a predominance of TcVI isolate in blood and tissues respect to TcIII and TcV; and a decrease of the inflammatory response in heart when the damage of mice infected with TcVI and TcIII + TcVI mixture were compared. In addition, simultaneous presence of two isolates in the same tissue was not detected. Our results show that biological interactions between isolates with different biological behaviors lead to changes in their biological properties. The occurrence of interactions among different genotypes of T. cruzi observed in our mouse model suggests that these phenomena could also occur in natural cycles in the Chaco Region.
Collapse
Affiliation(s)
- Paula G. Ragone
- Unidad de Epidemiología Molecular, Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- * E-mail:
| | - Cecilia Pérez Brandán
- Unidad de Epidemiología Molecular, Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Mercedes Monje Rumi
- Unidad de Epidemiología Molecular, Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Nicolás Tomasini
- Unidad de Epidemiología Molecular, Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Juan J. Lauthier
- Unidad de Epidemiología Molecular, Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Rubén O. Cimino
- Cátedra de Química Biológica, Facultad de Ciencias Naturales, Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Alejandro Uncos
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Federico Ramos
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Anahí M. Alberti D´Amato
- Unidad de Epidemiología Molecular, Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Miguel A. Basombrío
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| | - Patricio Diosque
- Unidad de Epidemiología Molecular, Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
- Instituto de Patología Experimental, CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Universidad Nacional de Salta, Salta-Capital, Argentina
| |
Collapse
|
20
|
Herrera CP, Licon MH, Nation CS, Jameson SB, Wesson DM. Genotype diversity of Trypanosoma cruzi in small rodents and Triatoma sanguisuga from a rural area in New Orleans, Louisiana. Parasit Vectors 2015; 8:123. [PMID: 25890064 PMCID: PMC4344744 DOI: 10.1186/s13071-015-0730-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/12/2015] [Indexed: 12/18/2022] Open
Abstract
Background Chagas disease is an anthropozoonosis caused by the protozoan parasite Trypanosoma cruzi that represents a major public health problem in Latin America. Although the United States is defined as non-endemic for Chagas disease due to the rarity of human cases, the presence of T. cruzi has now been amply demonstrated as enzootic in different regions of the south of the country from Georgia to California. In southeastern Louisiana, a high T. cruzi infection rate has been demonstrated in Triatoma sanguisuga, the local vector in this area. However, little is known about the role of small mammals in the wild and peridomestic transmission cycles. Methods This study focused on the molecular identification and genotyping of T. cruzi in both small rodents and T. sanguisuga from a rural area of New Orleans, Louisiana. DNA extractions were prepared from rodent heart, liver, spleen and skeletal muscle tissues and from cultures established from vector feces. T. cruzi infection was determined by standard PCR using primers specific for the minicircle variable region of the kinetoplastid DNA (kDNA) and the highly repetitive genomic satellite DNA (satDNA). Genotyping of discrete typing units (DTUs) was performed by amplification of mini-exon and 18S and 24Sα rRNA genes and subsequent sequence analysis. Results The DTUs TcI, TcIV and, for the first time, TcII, were identified in tissues of mice and rats naturally infected with T. cruzi captured in an area of New Orleans, close to the house where the first human case of Chagas disease was reported in Louisiana. The T. cruzi infection rate in 59 captured rodents was 76%. The frequencies of the detected DTUs in such mammals were TcI 82%, TcII 22% and TcIV 9%; 13% of all infections contained more than one DTU. Conclusions Our results indicate a probable presence of a considerably greater diversity in T. cruzi DTUs circulating in the southeastern United States than previously reported. Understanding T. cruzi transmission dynamics in sylvatic and peridomestic cycles in mammals and insect vectors will be crucial to estimating the risk of local, vector-borne transmission of T. cruzi to humans in the United States.
Collapse
Affiliation(s)
- Claudia P Herrera
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Rm. 1824, New Orleans, LA, 70112, USA.
| | - Meredith H Licon
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Rm. 1824, New Orleans, LA, 70112, USA.
| | - Catherine S Nation
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Rm. 1824, New Orleans, LA, 70112, USA.
| | - Samuel B Jameson
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Rm. 1824, New Orleans, LA, 70112, USA.
| | - Dawn M Wesson
- Department of Tropical Medicine, Vector-Borne Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal Street, Rm. 1824, New Orleans, LA, 70112, USA.
| |
Collapse
|
21
|
Human leucocyte antigen-G (HLA-G) and its murine functional homolog Qa2 in the Trypanosoma cruzi Infection. Mediators Inflamm 2015; 2015:595829. [PMID: 25688175 PMCID: PMC4320874 DOI: 10.1155/2015/595829] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/17/2014] [Indexed: 12/28/2022] Open
Abstract
Genetic susceptibility factors, parasite strain, and an adequate modulation of the immune system seem to be crucial for disease progression after Trypanosoma cruzi infection. HLA-G and its murine functional homolog Qa2 have well-recognized immunomodulatory properties. We evaluated the HLA-G 3′ untranslated region (3′UTR) polymorphic sites (associated with mRNA stability and target for microRNA binding) and HLA-G tissue expression (heart, colon, and esophagus) in patients presenting Chagas disease, stratified according to the major clinical variants. Further, we investigated the transcriptional levels of Qa2 and other pro- and anti-inflammatory genes in affected mouse tissues during T. cruzi experimental acute and early chronic infection induced by the CL strain. Chagas disease patients exhibited differential HLA-G 3′UTR susceptibility allele/genotype/haplotype patterns, according to the major clinical variant (digestive/cardiac/mixed/indeterminate). HLA-G constitutive expression on cardiac muscle and colonic cells was decreased in Chagasic tissues; however, no difference was observed for Chagasic and non-Chagasic esophagus tissues. The transcriptional levels of Qa2 and other anti and proinflammatory (CTLA-4, PDCD1, IL-10, INF-γ, and NOS-2) genes were induced only during the acute T. cruzi infection in BALB/c and C57BL/6 mice. We present several lines of evidence indicating the role of immunomodulatory genes and molecules in human and experimental T. cruzi infection.
Collapse
|
22
|
Basso B, Marini V. Experimental Chagas disease in Balb/c mice previously vaccinated with T. rangeli. II. The innate immune response shows immunological memory: reality or fiction? Immunobiology 2014; 220:428-36. [PMID: 25454810 DOI: 10.1016/j.imbio.2014.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 10/01/2014] [Accepted: 10/12/2014] [Indexed: 11/30/2022]
Abstract
Trypanosoma cruzi is a real challenge to the host's immune system, because it requires strong humoral and cellular immune response to remove circulating trypomastigote forms, and to prevent the replication of amastigote forms in tissues, involving many regulator and effector components. This protozoan is responsible for Chagas disease, a major public health problem in Latinamerica. We have developed a model of vaccination with Trypanosoma rangeli, a parasite closely related to T. cruzi, but nonpathogenic to humans, which reduces the infectiousness in three different species of animals, mice, dogs and guinea pigs, against challenge with T. cruzi. In a previous work, we demonstrated that mice vaccinated with T. rangeli showed important soluble mediators that stimulate phagocytic activity versus only infected groups. The aim of this work was to study the innate immune response in mice vaccinated or not with T. rangeli. Different population cells and some soluble mediators (cytokines) in peritoneal fluid and plasma in mice vaccinated-infected and only infected with T. cruzi were studied. In the first hours of challenge vaccinated mice showed an increase of macrophages, NK, granulocytes, and regulation of IL6, IFNγ, TNFα and IL10, with an increase of IL12, with respect to only infected mice. Furthermore an increase was observed of Li T, Li B responsible for adaptative response. Finally the findings showed that the innate immune response plays an important role in vaccinated mice for the early elimination of the parasites, complementary with the adaptative immune response, suggesting that vaccination with T. rangeli modulates the innate response, which develops some kind of immunological memory, recognizing shared antigens with T. cruzi. These results could contribute to the knowledge of new mechanisms which would have an important role in the immune response to Chagas disease.
Collapse
Affiliation(s)
- B Basso
- Department of Paediatrics, Neonatology Service, Medicine School, National Cordoba University, Argentina; National Co-ordination of Vector Control, Argentina.
| | - V Marini
- Department of Immunology Medicine School, Catholic University of Cordoba, Argentina
| |
Collapse
|
23
|
Ortiz S, Zulantay I, Apt W, Saavedra M, Solari A. Transferability of Trypanosoma cruzi from mixed human host infection to Triatoma infestans and from insects to axenic culture. Parasitol Int 2014; 64:33-6. [PMID: 25240699 DOI: 10.1016/j.parint.2014.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 08/21/2014] [Accepted: 09/11/2014] [Indexed: 10/24/2022]
Abstract
The etiologic agent of Chagas disease is Trypanosoma cruzi, a protozoan whose life cycle involves obligatory passage through vertebrate and invertebrate hosts in a series of stages. The aim of this study was to explore the transferability of mixed discrete typing units (DTUs) of T. cruzi present in chronic chagasic patients when passed through an invertebrate host during xenodiagnosis (XD) and then when transferred to axenic cultures to obtain T. cruzi isolates. DTUs of T. cruzi present in these two hosts and axenic cultures were identified by kDNA PCR amplification and subsequent hybridization with DTU-specific probes. Mixtures of Tc I, Tc II, Tc V and Tc VI DTUs were detected in blood samples. However as a result of XD and axenic cultures it was possible to identify mostly Tc V. We conclude that the transferability of an isolate of T.cruzi derived from mixed DTUs present in human blood depends upon the starved invertebrate host used for xenodiagnosis.
Collapse
Affiliation(s)
- Sylvia Ortiz
- Laboratorio de Biología Molecular de Parásitos, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Inés Zulantay
- Laboratorio de Parasitología Básico-Clínico, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Werner Apt
- Laboratorio de Parasitología Básico-Clínico, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Miguel Saavedra
- Laboratorio de Parasitología Básico-Clínico, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Aldo Solari
- Laboratorio de Biología Molecular de Parásitos, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
24
|
Nisimura LM, Estato V, de Souza EM, Reis PA, Lessa MA, Castro-Faria-Neto HC, Pereira MCDS, Tibiriçá E, Garzoni LR. Acute Chagas disease induces cerebral microvasculopathy in mice. PLoS Negl Trop Dis 2014; 8:e2998. [PMID: 25010691 PMCID: PMC4091872 DOI: 10.1371/journal.pntd.0002998] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 05/30/2014] [Indexed: 12/16/2022] Open
Abstract
Cardiomyopathy is the main clinical form of Chagas disease (CD); however, cerebral manifestations, such as meningoencephalitis, ischemic stroke and cognitive impairment, can also occur. The aim of the present study was to investigate functional microvascular alterations and oxidative stress in the brain of mice in acute CD. Acute CD was induced in Swiss Webster mice (SWM) with the Y strain of Trypanosoma cruzi (T. cruzi). Cerebral functional capillary density (the number of spontaneously perfused capillaries), leukocyte rolling and adhesion and the microvascular endothelial-dependent response were analyzed over a period of fifteen days using intravital video-microscopy. We also evaluated cerebral oxidative stress with the thiobarbituric acid reactive species TBARS method. Compared with the non-infected group, acute CD significantly induced cerebral functional microvascular alterations, including (i) functional capillary rarefaction, (ii) increased leukocyte rolling and adhesion, (iii) the formation of microvascular platelet-leukocyte aggregates, and (iv) alteration of the endothelial response to acetylcholine. Moreover, cerebral oxidative stress increased in infected animals. We concluded that acute CD in mice induced cerebral microvasculopathy, characterized by a reduced incidence of perfused capillaries, a high number of microvascular platelet-leukocyte aggregates, a marked increase in leukocyte-endothelium interactions and brain arteriolar endothelial dysfunction associated with oxidative stress. These results suggest the involvement of cerebral microcirculation alterations in the neurological manifestations of CD. Chagas disease (CD) is a neglected tropical illness caused by the parasite Trypanosoma cruzi (T. cruzi). It is endemic in Latin America and affects 10 million people worldwide. Meningoencephalitis occurs in children with acute CD and in immunosuppressed patients suffering acute CD reactivation. During the chronic phase, cerebral manifestations, including ischemic stroke and cognitive impairment, can also occur. Although microvascular alterations have been implicated in Chagas cardiomyopathy, the main clinical form of the disease, there is a lack of discussion in some studies regarding alterations of the cerebral microcirculation in CD. In the present study, we evaluated the functionality of the cerebral microcirculation in mice infected by T. cruzi. Utilizing an intravital video-microscope, we observed in the brain of infected mice a reduction in the number of perfused capillaries, an increased interaction between inflammatory cells and venules, the presence of microvascular platelet-leukocyte aggregates and alterations in the dilatation capacity of arterioles. Moreover, cerebral oxidative stress was increased in infected animals. We concluded that acute CD induced cerebral microvasculopathy.
Collapse
Affiliation(s)
- Lindice Mitie Nisimura
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Estato
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elen Mello de Souza
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia A. Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Adriano Lessa
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hugo Caire Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultra-estrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo Tibiriçá
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Ribeiro Garzoni
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: ,
| |
Collapse
|
25
|
Lewis MD, Fortes Francisco A, Taylor MC, Burrell-Saward H, McLatchie AP, Miles MA, Kelly JM. Bioluminescence imaging of chronic Trypanosoma cruzi infections reveals tissue-specific parasite dynamics and heart disease in the absence of locally persistent infection. Cell Microbiol 2014; 16:1285-300. [PMID: 24712539 PMCID: PMC4190689 DOI: 10.1111/cmi.12297] [Citation(s) in RCA: 178] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 03/18/2014] [Indexed: 12/12/2022]
Abstract
Summary Chronic Trypanosoma cruzi infections lead to cardiomyopathy in 20–30% of cases. A causal link between cardiac infection and pathology has been difficult to establish because of a lack of robust methods to detect scarce, focally distributed parasites within tissues. We developed a highly sensitive bioluminescence imaging system based on T. cruzi expressing a novel luciferase that emits tissue-penetrating orange-red light. This enabled long-term serial evaluation of parasite burdens in individual mice with an in vivo limit of detection of significantly less than 1000 parasites. Parasite distributions during chronic infections were highly focal and spatiotemporally dynamic, but did not localize to the heart. End-point ex vivo bioluminescence imaging allowed tissue-specific quantification of parasite loads with minimal sampling bias. During chronic infections, the gastro-intestinal tract, specifically the colon and stomach, was the only site where T. cruzi infection was consistently observed. Quantitative PCR-inferred parasite loads correlated with ex vivo bioluminescence and confirmed the gut as the parasite reservoir. Chronically infected mice developed myocarditis and cardiac fibrosis, despite the absence of locally persistent parasites. These data identify the gut as a permissive niche for long-term T. cruzi infection and show that canonical features of Chagas disease can occur without continual myocardium-specific infection.
Collapse
Affiliation(s)
- Michael D Lewis
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | | | | | | | | | | | | |
Collapse
|
26
|
Perez CJ, Lymbery AJ, Thompson RA. Chagas disease: the challenge of polyparasitism? Trends Parasitol 2014; 30:176-82. [DOI: 10.1016/j.pt.2014.01.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 01/31/2014] [Accepted: 01/31/2014] [Indexed: 01/19/2023]
|
27
|
Sanches TLM, Cunha LD, Silva GK, Guedes PMM, Silva JS, Zamboni DS. The use of a heterogeneously controlled mouse population reveals a significant correlation of acute phase parasitemia with mortality in Chagas disease. PLoS One 2014; 9:e91640. [PMID: 24651711 PMCID: PMC3961278 DOI: 10.1371/journal.pone.0091640] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/13/2014] [Indexed: 11/18/2022] Open
Abstract
Chagas disease develops upon infection with the protozoan parasite Trypanosoma cruzi and undergoes an acute phase characterized by massive parasite replication and the presence of parasites in the blood. This condition is known as acute phase parasitemia. This initial stage may result in a cure, in the development of the chronic stages of the disease or in the death of the infected host. Despite intensive investigation related to the characterization of the acute and chronic phases of the disease, the cause-effect relationship of acute phase parasitemia to the outcome of the disease is still poorly understood. In this study, we artificially generated a heterogeneously controlled mouse population by intercrossing F1 mice obtained from a parental breeding of highly susceptible A/J with highly resistant C57BL/6 mouse strains. This F2 population was infected and used to assess the correlation of acute phase parasitemia with the longevity of the animals. We used nonparametric statistical analyses and found a significant association between parasitemia and mortality. If males and females were evaluated separately, we found that the former were more susceptible to death, although parasitemia was similar in males and females. In females, we found a strong negative correlation between parasitemia and longevity. In males, however, additional factors independent of parasitemia may favor mouse mortality during the development of the disease. The correlations of acute phase parasitemia with mortality reported in this study may facilitate an appropriate prognostic approach to the disease in humans. Moreover, these results illustrate the complexity of the mammalian genetic traits that regulate host resistance during Chagas disease.
Collapse
Affiliation(s)
- Tiago L. M. Sanches
- Department of Cell Biology, University of São Paulo, Medical School Ribeirão Preto, FMRP/USP, São Paulo, Brazil
| | - Larissa D. Cunha
- Department of Cell Biology, University of São Paulo, Medical School Ribeirão Preto, FMRP/USP, São Paulo, Brazil
| | - Grace K. Silva
- Department of Biochemistry and Immunology, University of São Paulo, Medical School Ribeirão Preto, FMRP/USP, São Paulo, Brazil
| | - Paulo M. M. Guedes
- Department of Biochemistry and Immunology, University of São Paulo, Medical School Ribeirão Preto, FMRP/USP, São Paulo, Brazil
| | - João Santana Silva
- Department of Biochemistry and Immunology, University of São Paulo, Medical School Ribeirão Preto, FMRP/USP, São Paulo, Brazil
| | - Dario S. Zamboni
- Department of Cell Biology, University of São Paulo, Medical School Ribeirão Preto, FMRP/USP, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
28
|
Rodrigues GC, Feijó DF, Bozza MT, Pan P, Vullo D, Parkkila S, Supuran CT, Capasso C, Aguiar AP, Vermelho AB. Design, Synthesis, and Evaluation of Hydroxamic Acid Derivatives as Promising Agents for the Management of Chagas Disease. J Med Chem 2013; 57:298-308. [DOI: 10.1021/jm400902y] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Giseli Capaci Rodrigues
- Laboratório de Síntese Orgânica,
Departamento de Química, Instituto Militar de Engenharia, IME, Rio
de Janeiro, Brasil
- Laboratório Proteases
de Microrganismos, Departamento de Microbiologia, Instituto de Microbiologia Paulo de Góes, IMPG, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brasil
- Escola de
Ciência e Tecnologia e Programa de Pós-Graduação
em Ensino das Ciências, Universidade do Grande Rio, Unigranrio, Duque
de Caxias, Rio de Janeiro, Brasil
| | - Daniel Ferreira Feijó
- Laboratório de Inflamação e Imunidade,
Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, IMPPG, Universidade Federal do Rio de Janeiro, UFRJ, Rio
de Janeiro, Brasil
| | - Marcelo Torres Bozza
- Laboratório de Inflamação e Imunidade,
Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, IMPPG, Universidade Federal do Rio de Janeiro, UFRJ, Rio
de Janeiro, Brasil
| | - Peiwen Pan
- Institute of Biomedical
Technology, Fimlab Ltd., School of Medicine and BioMediTech, University of Tampere and Tampere University Hospital, Medisiinarinkatu 3, 33520 Tampere, Finland
| | - Daniela Vullo
- Laboratorio di Chimica Bioinorganica, Universita degli Studi di Firenze, Via della Lastruccia 3, Rm. 188, Polo Scientifico, 50019 Sesto Fiorentino, Florence, Italy
| | - Seppo Parkkila
- Institute of Biomedical
Technology, Fimlab Ltd., School of Medicine and BioMediTech, University of Tampere and Tampere University Hospital, Medisiinarinkatu 3, 33520 Tampere, Finland
| | - Claudiu T. Supuran
- Laboratorio di Chimica Bioinorganica, Universita degli Studi di Firenze, Via della Lastruccia 3, Rm. 188, Polo Scientifico, 50019 Sesto Fiorentino, Florence, Italy
- Dipartimento NEIROFARBA, Sezione di Scienze
Farmaceutiche, Universita degli Studi di Firenze, Via Ugo Schiff
6, 50019 Sesto Fiorentino, Florence, Italy
| | - Clemente Capasso
- Istituto di Biochimica delle Proteine, CNR, Via P. Castellino 111, 80131 Napoli, Italy
| | - Alcino Palermo Aguiar
- Laboratório de Síntese Orgânica,
Departamento de Química, Instituto Militar de Engenharia, IME, Rio
de Janeiro, Brasil
| | - Alane Beatriz Vermelho
- Laboratório Proteases
de Microrganismos, Departamento de Microbiologia, Instituto de Microbiologia Paulo de Góes, IMPG, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brasil
- Biotecnologia −
BIOINOVAR: Unidade de Bioenergia, Biocatalise e Bioprodutos, Instituto de Microbiologia Paulo de Góes, IMPG, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brasil
| |
Collapse
|
29
|
Sales-Campos H, Kappel HB, Andrade CP, Lima TP, Mattos ME, de Castilho A, Correia D, Giraldo LER, Lages-Silva E. A DTU-dependent blood parasitism and a DTU-independent tissue parasitism during mixed infection of Trypanosoma cruzi in immunosuppressed mice. Parasitol Res 2013; 113:375-85. [DOI: 10.1007/s00436-013-3665-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/18/2013] [Indexed: 01/09/2023]
|
30
|
Segatto M, Rodrigues CM, Machado CR, Franco GR, Pena SDJ, Macedo AM. LSSP-PCR of Trypanosoma cruzi: how the single primer sequence affects the kDNA signature. BMC Res Notes 2013; 6:174. [PMID: 23639061 PMCID: PMC3653686 DOI: 10.1186/1756-0500-6-174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 04/25/2013] [Indexed: 11/12/2022] Open
Abstract
Background Low-stringency single specific primer PCR (LSSP-PCR) is a highly sensitive and discriminating technique that has been extensively used to genetically characterize Trypanosoma cruzi populations in the presence of large amounts of host DNA. To ensure high sensitivity, in most T. cruzi studies, the variable regions of the naturally amplified kinetoplast DNA (kDNA) minicircles were targeted, and this method translated the intraspecific polymorphisms of these molecules into specific and reproducible kDNA signatures. Although the LSSP-PCR technique is reproducible under strict assay conditions, the complex banding pattern generated can be significantly altered by even a single-base change in the target DNA. Our survey of the literature identified eight different primers with similar, if not identical, names that have been used for kDNA amplification and LSSP-PCR of T. cruzi. Although different primer sequences were used in these studies, many of the authors cited the same reference report to justify their primer choice. We wondered whether these changes in the primer sequence could affect also the parasite LSSP-PCR profiles. Findings To answer this question we compared the kDNA signatures obtained from three different and extensively studied T. cruzi populations with the eight primers found in the literature. Our results clearly demonstrate that even minimal modifications in the oligonucleotide sequences, especially in the 3′ or 5′ end, can significantly change the kDNA signature of a T. cruzi strain. Conclusions These results highlight the necessity of careful preservation of primer nomenclature and sequence when reproducing an LSSP-PCR work to avoid confusion and allow comparison of results among different laboratories.
Collapse
Affiliation(s)
- Marcela Segatto
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | |
Collapse
|
31
|
Botero A, Thompson CK, Peacock CS, Clode PL, Nicholls PK, Wayne AF, Lymbery AJ, Thompson RCA. Trypanosomes genetic diversity, polyparasitism and the population decline of the critically endangered Australian marsupial, the brush tailed bettong or woylie (Bettongia penicillata). INTERNATIONAL JOURNAL FOR PARASITOLOGY-PARASITES AND WILDLIFE 2013; 2:77-89. [PMID: 24533319 PMCID: PMC3862532 DOI: 10.1016/j.ijppaw.2013.03.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 03/13/2013] [Accepted: 03/14/2013] [Indexed: 12/14/2022]
Abstract
Trypanosomes in Australian marsupials comprise a heterogeneous community. The woylie was the only species found co-infected with different trypanosomes. Some of the trypanosomes found are able to colonize several tissues in the host. Trypanosoma copemani is able to invade cells in vitro. Association between T.copemani and mixed infections with the decline of the woylie.
While much is known of the impact of trypanosomes on human and livestock health, trypanosomes in wildlife, although ubiquitous, have largely been considered to be non-pathogenic. We describe the genetic diversity, tissue tropism and potential pathogenicity of trypanosomes naturally infecting Western Australian marsupials. Blood samples collected from 554 live-animals and 250 tissue samples extracted from 50 carcasses of sick-euthanized or road-killed animals, belonging to 10 species of marsupials, were screened for the presence of trypanosomes using a PCR of the 18S rDNA gene. PCR results revealed a rate of infection of 67% in blood and 60% in tissues. Inferred phylogenetic trees using 18S rDNA and glycosomal glyceraldehyde phosphate dehydrogenase (gGAPDH) sequences showed the presence of eight genotypes that clustered into three clades: a clade including Trypanosoma copemani, a new clade closely related to Trypanosoma gilletti, and a clade including Trypanosoma H25 from an Australian kangaroo. Trypanosome infections were compared in a declining and in a stable population of the endangered Australian marsupial, the brush tailed bettong or woylie (Bettongia penicillata). This marsupial showed high rates of infection with Clade A genotypes (96%) in the declining population, whereas in the stable population, Clade B genotypes were predominant (89%). Mixed infections were common in woylies from the declining but not from the stable population. Histopathological findings associated with either mixed or single infections involving Clade A genotypes, showed a strong inflammatory process and tissue degeneration predominantly in heart, oesophagus and tongue. Trypanosomes were successfully grown in culture and for the first time we demonstrate that a genotype within Clade A has the capacity to not only colonize different tissues in the host but also to invade cells in vitro. These results provide evidence for the potential role of trypanosomes in the decline of a formerly abundant marsupial that is now critically endangered.
Collapse
Affiliation(s)
- Adriana Botero
- School of Veterinary and Life Sciences, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
| | - Craig K Thompson
- School of Veterinary and Life Sciences, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
| | - Christopher S Peacock
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, WA 6009, Australia ; Telethon Institute for Child Health Research, 100 Roberts Road, Subiaco, WA 6008, Australia
| | - Peta L Clode
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Stirling HWY, Crawley, WA 6009, Australia
| | - Philip K Nicholls
- School of Veterinary and Life Sciences, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
| | - Adrian F Wayne
- Department of Environment and Conservation, Science Division, Manjimup, WA, Australia
| | - Alan J Lymbery
- Fish Health Unit, School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch, WA 6150, Australia
| | - R C Andrew Thompson
- School of Veterinary and Life Sciences, Murdoch University, 90 South Street, Murdoch, WA 6150, Australia
| |
Collapse
|
32
|
Hashimoto M, Enomoto M, Morales J, Kurebayashi N, Sakurai T, Hashimoto T, Nara T, Mikoshiba K. Inositol 1,4,5-trisphosphate receptor regulates replication, differentiation, infectivity and virulence of the parasitic protist Trypanosoma cruzi. Mol Microbiol 2013; 87:1133-50. [PMID: 23320762 DOI: 10.1111/mmi.12155] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2013] [Indexed: 11/26/2022]
Abstract
In animals, inositol 1,4,5-trisphosphate receptors (IP3 Rs) are ion channels that play a pivotal role in many biological processes by mediating Ca(2+) release from the endoplasmic reticulum. Here, we report the identification and characterization of a novel IP3 R in the parasitic protist, Trypanosoma cruzi, the pathogen responsible for Chagas disease. DT40 cells lacking endogenous IP3 R genes expressing T. cruzi IP3 R (TcIP3 R) exhibited IP3 -mediated Ca(2+) release from the ER, and demonstrated receptor binding to IP3 . TcIP3 R was expressed throughout the parasite life cycle but the expression level was much lower in bloodstream trypomastigotes than in intracellular amastigotes or epimastigotes. Disruption of two of the three TcIP3 R gene loci led to the death of the parasite, suggesting that IP3 R is essential for T. cruzi. Parasites expressing reduced or increased levels of TcIP3 R displayed defects in growth, transformation and infectivity, indicating that TcIP3 R is an important regulator of the parasite's life cycle. Furthermore, mice infected with T. cruzi expressing reduced levels of TcIP3 R exhibited a reduction of disease symptoms, indicating that TcIP3 R is an important virulence factor. Combined with the fact that the primary structure of TcIP3 R has low similarity to that of mammalian IP3 Rs, TcIP3 R is a promising drug target for Chagas disease.
Collapse
Affiliation(s)
- Muneaki Hashimoto
- Department of Molecular and Cellular Parasitology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Andrade SG, Campos RF, Steindel M, Guerreiro ML, Magalhães JB, Almeida MCD, Reis JN, Santos VC, Valadares HMS, Reis MGD, Macedo AM. Biological, biochemical and molecular features of Trypanosoma cruzi strains isolated from patients infected through oral transmission during a 2005 outbreak in the state of Santa Catarina, Brazil: its correspondence with the new T. cruzi Taxonomy Consensus (2009). Mem Inst Oswaldo Cruz 2012; 106:948-56. [PMID: 22241116 DOI: 10.1590/s0074-02762011000800009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 09/22/2011] [Indexed: 11/22/2022] Open
Abstract
We examined strains of Trypanosoma cruzi isolated from patients with acute Chagas disease that had been acquired by oral transmission in the state of Santa Catarina, Brazil (2005) and two isolates that had been obtained from a marsupial (Didelphis aurita) and a vector (Triatoma tibiamaculata). These strains were characterised through their biological behaviour and isoenzymic profiles and genotyped according to the new Taxonomy Consensus (2009) based on the discrete typing unities, that is, T. cruzi genotypes I-VI. All strains exhibited the biological behaviour of biodeme type II. In six isolates, late peaks of parasitaemia, beyond the 20th day, suggested a double infection with biodemes II + III. Isoenzymes revealed Z2 or mixed Z1 and Z2 profiles. Genotyping was performed using three polymorphic genes (cytochrome oxidase II, spliced leader intergenic region and 24Sα rRNA) and the restriction fragment length polymorphism of the kDNA minicircles. Based on these markers, all but four isolates were characterised as T. cruzi II genotypes. Four mixed populations were identified: SC90, SC93 and SC97 (T. cruzi I + T. cruzi II) and SC95 (T. cruzi I + T. cruzi VI). Comparison of the results obtained by different methods was essential for the correct identification of the mixed populations and major lineages involved indicating that characterisation by different methods can provide new insights into the relationship between phenotypic and genotypic aspects of parasite behaviour.
Collapse
Affiliation(s)
- Sonia Gumes Andrade
- Laboratório de Chagas Experimental, Autoimunidade e Imunologia Celular, Centro de Pesquisas Gonçalo Moniz, Fiocruz, Salvador, BA, Brasil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Duell BL, Tan CK, Carey AJ, Wu F, Cripps AW, Ulett GC. Recent insights into microbial triggers of interleukin-10 production in the host and the impact on infectious disease pathogenesis. ACTA ACUST UNITED AC 2012; 64:295-313. [PMID: 22268692 DOI: 10.1111/j.1574-695x.2012.00931.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 01/17/2012] [Accepted: 01/17/2012] [Indexed: 02/06/2023]
Abstract
Since its initial description as a Th2-cytokine antagonistic to interferon-alpha and granulocyte-macrophage colony-stimulating factor, many studies have shown various anti-inflammatory actions of interleukin-10 (IL-10), and its role in infection as a key regulator of innate immunity. Studies have shown that IL-10 induced in response to microorganisms and their products plays a central role in shaping pathogenesis. IL-10 appears to function as both sword and shield in the response to varied groups of microorganisms in its capacity to mediate protective immunity against some organisms but increase susceptibility to other infections. The nature of IL-10 as a pleiotropic modulator of host responses to microorganisms is explained, in part, by its potent and varied effects on different immune effector cells which influence antimicrobial activity. A new understanding of how microorganisms trigger IL-10 responses is emerging, along with recent discoveries of how IL-10 produced during disease might be harnessed for better protective or therapeutic strategies. In this review, we summarize studies from the past 5 years that have reported the induction of IL-10 by different classes of pathogenic microorganisms, including protozoa, nematodes, fungi, viruses and bacteria and discuss the impact of this induction on the persistence and/or clearance of microorganisms in the host.
Collapse
Affiliation(s)
- Benjamin L Duell
- School of Medical Sciences, Centre for Medicine and Oral Health, Griffith University, Gold Coast Campus, Gold Coast, Qld, Australia
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Trypanosomes are protozoan parasites of medical and veterinary importance. It is well established that different species, subspecies and strains of trypanosome can cause very different disease in the mammalian host, exemplified by the two human-infective subspecies of Trypanosoma brucei that cause either acute or chronic disease. We are beginning to understand how the host response shapes the course of the disease and how genetic variation in the host can be a factor in disease severity, particularly in the mouse model, but until recently the role of parasite genetic variation that determines differential disease outcome has been a neglected area. This review will discuss the recent advances in this field, covering both our current knowledge of the T. brucei genes involved and the approaches that are leading towards the identification of T. brucei virulence genes. Finally, the potential for using parasite genotype variation to examine the evolutionary context of virulence will be discussed.
Collapse
Affiliation(s)
- L J Morrison
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
36
|
Terrazas CA, Huitron E, Vazquez A, Juarez I, Camacho GM, Calleja EA, Rodriguez-Sosa M. MIF synergizes with Trypanosoma cruzi antigens to promote efficient dendritic cell maturation and IL-12 production via p38 MAPK. Int J Biol Sci 2011; 7:1298-310. [PMID: 22110382 PMCID: PMC3221366 DOI: 10.7150/ijbs.7.1298] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 01/30/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) has been found to be involved in host resistance to several parasitic infections. To determine the mechanisms of the MIF-dependent responses to Trypanosoma cruzi, we investigated host resistance in MIF-/- mice (on the BALB/c background) during an intraperitoneal infection. We focused on the potential involvement of MIF in dendritic cell (DC) maturation and cytokine production. Following a challenge with 5 x 103T. cruzi parasites, wild type (WT) mice developed a strong IL-12 response and adequate maturation of the draining mesenteric lymph node DCs and were resistant to infection. In contrast, similarly infected MIF-/- mice mounted a weak IL-12 response, displayed immature DCs in the early phases of infection and rapidly succumbed to T. cruzi infection. The lack of maturation and IL-12 production by the DCs in response to total T. cruzi antigen (TcAg) was confirmed by in vitro studies. These effects were reversed following treatment with recombinant MIF. Interestingly, TcAg-stimulated bone marrow-derived DCs from both WT and MIF-/- mice had increased ERK1/2 MAPK phosphorylation. In contrast, p38 phosphorylation was only upregulated in WT DCs. Reconstitution of MIF to MIF-/- DCs upregulated p38 phosphorylation. The MIF-p38 pathway affected MHC-II and CD86 expression as well as IL-12 production. These findings demonstrate that the MIF-induced early DC maturation and IL-12 production mediates resistance to T. cruzi infection, probably by activating the p38 pathway.
Collapse
Affiliation(s)
- Cesar A Terrazas
- Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México-UNAM, C. P. 54090 Estado de México, Mexico
| | | | | | | | | | | | | |
Collapse
|
37
|
DosReis GA. Evasion of immune responses by Trypanosoma cruzi, the etiological agent of Chagas disease. Braz J Med Biol Res 2011; 44:84-90. [PMID: 21243314 DOI: 10.1590/s0100-879x2011007500005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 01/03/2011] [Indexed: 01/02/2023] Open
Abstract
Infection with the protozoan parasite Trypanosoma cruzi leads to Chagas disease, which affects millions of people in Latin America. Infection with T. cruzi cannot be eliminated by the immune system. A better understanding of immune evasion mechanisms is required in order to develop more effective vaccines. During the acute phase, parasites replicate extensively and release immunomodulatory molecules that delay parasite-specific responses mediated by T cells. This immune evasion allows the parasite to spread in the host. In the chronic phase, parasite evasion relies on its replication strategy of hijacking the TGF-β signaling pathway involved in inflammation and tissue regeneration. In this article, the mechanisms of immune evasion described for T. cruzi are reviewed.
Collapse
Affiliation(s)
- G A DosReis
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|