1
|
Rigazio CS, Mariz-Ponte N, Caballero EP, Penas FN, Goren NB, Santamaría MH, Corral RS. Involvement of glycoinositolphospholipid from Trypanosoma cruzi and macrophage migration inhibitory factor in proinflammatory mechanisms promoting cardiovascular injury mechanisms promoting cardiovascular inflammation tThe combined action of glycoinositolphospholipid from Trypanosoma cruzi and macrophage migration inhibitory factor increases proinflammatory mediator production by cardiomyocytes and vascular endothelial cells. Microb Pathog 2022; 173:105881. [DOI: 10.1016/j.micpath.2022.105881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022]
|
2
|
Barbosa CHD, Canto FB, Gomes A, Brandao LM, Lima JR, Melo GA, Granato A, Neves EGA, Dutra WO, Oliveira AC, Nóbrega A, Bellio M. Cytotoxic CD4+ T cells driven by T-cell intrinsic IL-18R/MyD88 signaling predominantly infiltrate Trypanosoma cruzi-infected hearts. eLife 2022; 11:74636. [PMID: 35670567 PMCID: PMC9236613 DOI: 10.7554/elife.74636] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 06/04/2022] [Indexed: 11/13/2022] Open
Abstract
Increasing attention has been directed to cytotoxic CD4+ T cells (CD4CTLs) in different pathologies, both in humans and mice. The impact of CD4CTLs in immunity and the mechanisms controlling their generation, however, remain poorly understood. Here, we show that CD4CTLs abundantly differentiate during mouse infection with the intracellular parasite Trypanosoma cruzi. CD4CTLs display parallel kinetics to Th1 cells in the spleen, mediate specific cytotoxicity against cells presenting pathogen-derived antigens and express immunoregulatory and/or exhaustion markers. We demonstrate that CD4CTL absolute numbers and activity are severely reduced in both Myd88-/- and Il18ra-/- mice. Of note, the infection of mixed-bone marrow chimeras revealed that WT but not Myd88-/- cells transcribe the CD4CTL gene signature and that Il18ra-/- and Myd88-/- CD4+ T cells phenocopy each other. Moreover, adoptive transfer of WT CD4+GzB+ T cells to infected Il18ra-/- mice extended their survival. Importantly, cells expressing the CD4CTL phenotype predominate among CD4+ T cells infiltrating the infected mouse cardiac tissue and are increased in the blood of Chagas patients, in which the frequency of CD4CTLs correlates with the severity of cardiomyopathy. Our findings describe CD4CTLs as a major player in immunity to a relevant human pathogen and disclose T-cell intrinsic IL-18R/MyD88 signaling as a key pathway controlling the magnitude of the CD4CTL response.
Collapse
Affiliation(s)
| | - Fabio B Canto
- Departamento de Imunobiologia, Universidade Federal Fluminense
| | - Ariel Gomes
- Department of Immunology, Universidade Federal do Rio de Janeiro (UFRJ)
| | - Layza M Brandao
- Department of Immunology, Universidade Federal do Rio de Janeiro (UFRJ)
| | - Jéssica R Lima
- Department of Immunology, Universidade Federal do Rio de Janeiro (UFRJ)
| | - Guilherme A Melo
- Department of Immunology, Universidade Federal do Rio de Janeiro (UFRJ)
| | | | - Eula GA Neves
- Laboratório de Biologia das Interações Celulares, Universidade Federal de Minas Gerais
| | - Walderez O Dutra
- Laboratório de Biologia das Interações Celulares, Universidade Federal de Minas Gerais
| | - Ana-Carolina Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ)
| | - Alberto Nóbrega
- Department of Immunology, Universidade Federal do Rio de Janeiro (UFRJ)
| | - Maria Bellio
- Department of Immunology, Universidade Federal do Rio de Janeiro (UFRJ)
| |
Collapse
|
3
|
Borges AR, Link F, Engstler M, Jones NG. The Glycosylphosphatidylinositol Anchor: A Linchpin for Cell Surface Versatility of Trypanosomatids. Front Cell Dev Biol 2021; 9:720536. [PMID: 34790656 PMCID: PMC8591177 DOI: 10.3389/fcell.2021.720536] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/06/2021] [Indexed: 11/20/2022] Open
Abstract
The use of glycosylphosphatidylinositol (GPI) to anchor proteins to the cell surface is widespread among eukaryotes. The GPI-anchor is covalently attached to the C-terminus of a protein and mediates the protein’s attachment to the outer leaflet of the lipid bilayer. GPI-anchored proteins have a wide range of functions, including acting as receptors, transporters, and adhesion molecules. In unicellular eukaryotic parasites, abundantly expressed GPI-anchored proteins are major virulence factors, which support infection and survival within distinct host environments. While, for example, the variant surface glycoprotein (VSG) is the major component of the cell surface of the bloodstream form of African trypanosomes, procyclin is the most abundant protein of the procyclic form which is found in the invertebrate host, the tsetse fly vector. Trypanosoma cruzi, on the other hand, expresses a variety of GPI-anchored molecules on their cell surface, such as mucins, that interact with their hosts. The latter is also true for Leishmania, which use GPI anchors to display, amongst others, lipophosphoglycans on their surface. Clearly, GPI-anchoring is a common feature in trypanosomatids and the fact that it has been maintained throughout eukaryote evolution indicates its adaptive value. Here, we explore and discuss GPI anchors as universal evolutionary building blocks that support the great variety of surface molecules of trypanosomatids.
Collapse
Affiliation(s)
- Alyssa R Borges
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Fabian Link
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Markus Engstler
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Nicola G Jones
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Rodrigues da Cunha GM, Azevedo MA, Nogueira DS, Clímaco MDC, Valencia Ayala E, Jimenez Chunga JA, La Valle RJY, da Cunha Galvão LM, Chiari E, Brito CRN, Soares RP, Nogueira PM, Fujiwara RT, Gazzinelli R, Hincapie R, Chaves CS, Oliveira FMS, Finn MG, Marques AF. α-Gal immunization positively impacts Trypanosoma cruzi colonization of heart tissue in a mouse model. PLoS Negl Trop Dis 2021; 15:e0009613. [PMID: 34314435 PMCID: PMC8345864 DOI: 10.1371/journal.pntd.0009613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/06/2021] [Accepted: 06/30/2021] [Indexed: 01/03/2023] Open
Abstract
Chagas disease, caused by the parasite Trypanosoma cruzi, is considered endemic in more than 20 countries but lacks both an approved vaccine and limited treatment for its chronic stage. Chronic infection is most harmful to human health because of long-term parasitic infection of the heart. Here we show that immunization with a virus-like particle vaccine displaying a high density of the immunogenic α-Gal trisaccharide (Qβ-αGal) induced several beneficial effects concerning acute and chronic T. cruzi infection in α1,3-galactosyltransferase knockout mice. Approximately 60% of these animals were protected from initial infection with high parasite loads. Vaccinated animals also produced high anti-αGal IgG antibody titers, improved IFN-γ and IL-12 cytokine production, and controlled parasitemia in the acute phase at 8 days post-infection (dpi) for the Y strain and 22 dpi for the Colombian strain. In the chronic stage of infection (36 and 190 dpi, respectively), all of the vaccinated group survived, showing significantly decreased heart inflammation and clearance of amastigote nests from the heart tissue.
Collapse
Affiliation(s)
| | - Maíra Araújo Azevedo
- Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, Brazil
| | - Denise Silva Nogueira
- Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, Brazil
| | | | | | - Juan Atilio Jimenez Chunga
- Universidad Nacional Mayor de San Marcos, Faculdad de Ciencias Biologicas, Escuela Profesional de Microbiología y Parasitología—Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Lima, Peru
| | - Raul Jesus Ynocente La Valle
- Universidad Nacional Mayor de San Marcos, Faculdad de Ciencias Biologicas, Escuela Profesional de Microbiología y Parasitología—Laboratorio de Parasitología en Fauna Silvestre y Zoonosis, Lima, Peru
| | | | - Egler Chiari
- Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, Brazil
| | - Carlos Ramon Nascimento Brito
- Universidade Federal do Rio Grande do Norte—Centro de Ciências da Saúde—Departamento de Análises Clínicas e Toxicológicas, Natal, Brazil
| | | | | | | | - Ricardo Gazzinelli
- Universidade Federal de Minas Gerais, Departamento de Parasitologia, Belo Horizonte, Brazil
- Instituto René Rachou/FIOCRUZ–MG, Belo Horizonte, Brazil
| | - Robert Hincapie
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Carlos-Sanhueza Chaves
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | | | - M. G. Finn
- School of Chemistry and Biochemistry, School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | | |
Collapse
|
5
|
Gil-Jaramillo N, Rocha AP, Raiol T, Motta FN, Favali C, Brigido MM, Bastos IMD, Santana JM. The First Contact of Human Dendritic Cells With Trypanosoma cruzi Reveals Response to Virus as an Unexplored Central Pathway. Front Immunol 2021; 12:638020. [PMID: 33897690 PMCID: PMC8062726 DOI: 10.3389/fimmu.2021.638020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/11/2021] [Indexed: 12/21/2022] Open
Abstract
Chagas disease is a debilitating and neglected disease caused by the protozoan Trypanosoma cruzi. Soon after infection, interactions among T. cruzi and host innate immunity cells can drive/contribute to disease outcome. Dendritic cells (DCs), present in all tissues, are one of the first immune cells to interact with Trypanosoma cruzi metacyclic trypomastigotes. Elucidating the immunological events triggered immediately after parasite-human DCs encounter may aid in understanding the role of DCs in the establishment of infection and in the course of the disease. Therefore, we performed a transcriptomic analysis of a 12 h interaction between T. cruzi and MoDCs (monocyte-derived DCs) from three human donors. Enrichment analyses of the 468 differentially expressed genes (DEGs) revealed viral infection response as the most regulated pathway. Additionally, exogenous antigen processing and presentation through MHC-I, chemokine signaling, lymphocyte co-stimulation, metallothioneins, and inflammasome activation were found up-regulated. Notable, we were able to identify the increased gene expression of alternative inflammasome sensors such as AIM2, IFI16, and RIG-I for the first time in a T. cruzi infection. Both transcript and protein expression levels suggest proinflammatory cytokine production during early T. cruzi-DCs contact. Our transcriptome data unveil antiviral pathways as an unexplored process during T. cruzi-DC initial interaction, disclosing a new panorama for the study of Chagas disease outcomes.
Collapse
Affiliation(s)
- Natalia Gil-Jaramillo
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Amanda Pereira Rocha
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Tainá Raiol
- Fiocruz Brasília–Gerência Regional de Brasília (GEREB), Fundação Oswaldo Cruz (Fiocruz), Brasília, Brazil
| | - Flávia Nader Motta
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
- Faculdade de Ceilândia, Universidade de Brasília, Brasília, Brazil
| | - Cecília Favali
- Laboratório de Imunologia Celular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Marcelo M. Brigido
- Laboratório de Imunologia Molecular, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Izabela M. D. Bastos
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| | - Jaime M. Santana
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
6
|
Zheng W, Xu Q, Zhang Y, E X, Gao W, Zhang M, Zhai W, Rajkumar RS, Liu Z. Toll-like receptor-mediated innate immunity against herpesviridae infection: a current perspective on viral infection signaling pathways. Virol J 2020; 17:192. [PMID: 33298111 PMCID: PMC7726878 DOI: 10.1186/s12985-020-01463-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Background In the past decades, researchers have demonstrated the critical role of Toll-like receptors (TLRs) in the innate immune system. They recognize viral components and trigger immune signal cascades to subsequently promote the activation of the immune system. Main body Herpesviridae family members trigger TLRs to elicit cytokines in the process of infection to activate antiviral innate immune responses in host cells. This review aims to clarify the role of TLRs in the innate immunity defense against herpesviridae, and systematically describes the processes of TLR actions and herpesviridae recognition as well as the signal transduction pathways involved. Conclusions Future studies of the interactions between TLRs and herpesviridae infections, especially the subsequent signaling pathways, will not only contribute to the planning of effective antiviral therapies but also provide new molecular targets for the development of antiviral drugs.
Collapse
Affiliation(s)
- Wenjin Zheng
- School of Basic Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Qing Xu
- School of Anesthesiology, Weifang Medical University, Weifang, 261053, China
| | - Yiyuan Zhang
- School of Basic Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Xiaofei E
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Wei Gao
- Key Lab for Immunology in Universities of Shandong Province, School of Basic Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Mogen Zhang
- School of Basic Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | - Weijie Zhai
- School of Basic Medical Sciences, Weifang Medical University, Weifang, 261053, China
| | | | - Zhijun Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Weifang Medical University, Weifang, 261053, China.
| |
Collapse
|
7
|
Costa GC, Rocha MODC, Souza PEAD, Melo DFS, Moreira PR, Gollob KJ, Nunes MDCP, Dutra WO. CD14 genotype and functional dichotomy of CD14+ and CD14- cells are associated with activated immune response and development of Chagas dilated cardiomyopathy. Mem Inst Oswaldo Cruz 2020; 115:e200110. [PMID: 33146244 PMCID: PMC7592494 DOI: 10.1590/0074-02760200110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/24/2020] [Indexed: 11/22/2022] Open
Abstract
We aimed to investigate the association of CD14 -260C/T (rs2569190) polymorphism and Chagas cardiomyopathy and the functional characteristics of CD14+ and CD14- monocytes upon infection with Trypanosoma cruzi. We observed an association between the T- genotype (absence of allele -260T) related to low CD14 expression and the dilated cardiomyopathy type of Chagas disease. Furthermore, we observed that CD14- monocytes showed a more activated profile upon in vitro infection with T. cruzi than CD14+ monocytes. Our findings suggest that T- genotype is associated with susceptibility to develop Chagas dilated cardiomyopathy, likely linked to the T. cruzi-induced inflammatory profile of CD14- monocytes.
Collapse
Affiliation(s)
| | - Manoel Otávio da Costa Rocha
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Infectologia e Medicina Tropical, Belo Horizonte, MG, Brasil
| | | | - Diego Felipe Sa Melo
- Universidade Federal de Minas Gerais, Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Belo Horizonte, MG, Brasil
| | - Paula Rocha Moreira
- Universidade Federal de Minas Gerais, Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Belo Horizonte, MG, Brasil
| | - Kenneth John Gollob
- AC Camargo Cancer Center, São Paulo, SP, Brasil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, Salvador, BA, Brasil
| | - Maria do Carmo Pereira Nunes
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Infectologia e Medicina Tropical, Belo Horizonte, MG, Brasil
| | - Walderez Ornelas Dutra
- Universidade Federal de Minas Gerais, Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Belo Horizonte, MG, Brasil.,Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, Salvador, BA, Brasil
| |
Collapse
|
8
|
Mohammed ASA, Tian W, Zhang Y, Peng P, Wang F, Li T. Leishmania lipophosphoglycan components: A potent target for synthetic neoglycoproteins as a vaccine candidate for leishmaniasis. Carbohydr Polym 2020; 237:116120. [PMID: 32241437 DOI: 10.1016/j.carbpol.2020.116120] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 11/27/2022]
Abstract
Leishmania is an obligate intracellular pathogen that invades phagocytic host cells. Due to its high morbidity and mortality rates, leishmaniasis attracts significant attention. The disease, which is caused by Leishmania parasites, is distributed worldwide, particularly among developing communities, and causes fatal complications if not treated expediently. Unfortunately, the existing treatments are not preventive and do not impede Leishmania infection. Many drugs available for leishmaniasis are becoming less effective due to emerging resistance in some Leishmania species. Other drugs have drawbacks such as low cost-effectiveness, toxicity, and side effects. The World Health Organization (WHO) considers leishmaniasis to be a major public health problem and suggests that the best prevention is to develop a vaccine for this dangerous disease. In this review, we focus on the unique components of lipophosphoglycan (LPG), a component of the Leishmania cell wall, particularly [Galp(1 → 4)-β-[Manp-(1 → 2)-α-Manp-(1 → 2)-α]-Manp] in the cryptic tetrasaccharide cap, and on synthetic approaches as a potent candidate for a leishmaniasis vaccine.
Collapse
Affiliation(s)
- Aiman Saleh A Mohammed
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China; National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China
| | - Weilu Tian
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Youqin Zhang
- National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China
| | - Peng Peng
- National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China; National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China.
| | - Tianlu Li
- Key Laboratory of Chemical Biology (Ministry of Education), Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China; National Glycoengineering Research Center, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
9
|
Lonien SCH, Malvezi AD, Suzukawa HT, Yamauchi LM, Yamada-Ogatta SF, Rizzo LV, Bordignon J, Pinge-Filho P. Response to Trypanosoma cruzi by Human Blood Cells Enriched with Dentritic Cells Is Controlled by Cyclooxygenase-2 Pathway. Front Microbiol 2017; 8:2020. [PMID: 29118740 PMCID: PMC5660982 DOI: 10.3389/fmicb.2017.02020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 10/03/2017] [Indexed: 12/14/2022] Open
Abstract
Chagas disease (Cd) or American human trypanosomiasis is caused by Trypanosoma cruzi and affects ~7 million people, mostly in Latin America. The infective trypomastigote forms of the parasite can invade several human blood cell populations, including monocytes and dendritic cells (DC). Although these cells display a wide functional diversity, their interactions with T. cruzi via cyclooxygenase (COX) and cyclic adenosine monophosphate (cAMP) dependent pathways have not been analyzed. To exploiting this mechanism, DC-enriched peripheral human blood mononuclear cell populations (DC-PBMC) were used as our model. Our results showed that the treatment of these cell populations with celecoxib (CEL), a cyclooxygenase-2 selective inhibitor or SQ 22,536, an adenilate cyclase inhibitor, significantly caused marked inhibition of T. cruzi infection. In contrast, aspirin (ASA, a non-selective COX-1 and COX-2 inhibitor) treatment did not inhibit the infection of the cells by the parasite and was independent of nitric oxide (NO) production. The expression of co-stimulatory molecules CD80 and CD86 were similar on cells treated or not with both COX-inhibitors. The infection stimulated the release of TNF-α, IL-1β, IL-6, IL-8, and IL-10 production by infected cells. Treatment with ASA or CEL did not affect TNF-α, IL-6, IL-8, IL-10, and NO production by infected cells, but increased IL-1β production by them. Our results suggest a key role of COX-2 and cAMP pathways in T. cruzi invasion process of human blood cells and these pathways may represent targets of new therapeutic options for Cd.
Collapse
Affiliation(s)
- Sandra C H Lonien
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Aparecida D Malvezi
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Helena T Suzukawa
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Lucy M Yamauchi
- Laboratório de Biologia Molecular de Microrganismos, Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Sueli F Yamada-Ogatta
- Laboratório de Biologia Molecular de Microrganismos, Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Luiz V Rizzo
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Juliano Bordignon
- Laboratório de Virologia Molecular, Instituto Carlos Chagas/Fiocruz, Curitiba, Brazil
| | - Phileno Pinge-Filho
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
10
|
Passos LSA, Magalhães LMD, Soares RP, Marques AF, Nunes MDCP, Gollob KJ, Dutra WO. Specific activation of CD4 - CD8 - double-negative T cells by Trypanosoma cruzi-derived glycolipids induces a proinflammatory profile associated with cardiomyopathy in Chagas patients. Clin Exp Immunol 2017; 190:122-132. [PMID: 28543170 DOI: 10.1111/cei.12992] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2017] [Indexed: 12/13/2022] Open
Abstract
Cardiomyopathy is the most severe outcome of Chagas disease, causing more than 12 000 deaths/year. Immune cells participate in cardiomyopathy development either by direct tissue destruction, or by driving inflammation. We have shown that CD4- CD8- [double-negative (DN)] T cells are major sources of inflammatory and anti-inflammatory cytokines, associated with the cardiac (CARD) and indeterminate (IND) forms of Chagas disease, respectively. Here, we sought to identify Trypanosoma cruzi-derived components that lead to activation of DN T cells in Chagas patients. Glycolipid (GCL), lipid (LIP) and protein-enriched (PRO) fractions derived from trypomastigote forms of T. cruzi were utilized to stimulate cells from IND and CARD patients to determine DN T cell activation by evaluating CD69 and cytokine expression. We observed that GCL, but not LIP or PRO fractions, induced higher activation of DN T cells, especially T cell receptor (TCR)-γδ DN T, from IND and CARD. GCL led to an increase in tumour necrosis factor (TNF) and interleukin (IL)-10 expression by TCR-γδ DN T cells from IND, while inducing IFN-γ expression by TCR-γδ DN T cells from CARD. This led to an increase in the ratio IFN-γ/IL-10 in TCR-γδ DN T cells from CARD, favouring an inflammatory profile. These results identify GCL as the major T. cruzi component responsible for activation of DN T cells in chronic Chagas disease, associated predominantly with an inflammatory profile in CARD, but not IND. These findings may have implications for designing new strategies of control or prevention of Chagas disease cardiomyopathy by modulating the response to GCL.
Collapse
Affiliation(s)
- L S A Passos
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,Pós-graduação em Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - L M D Magalhães
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,Pós-graduação em Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - R P Soares
- Pós-graduação em Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, FIOCRUZ, Belo Horizonte, MG, Brazil
| | - A F Marques
- Pós-graduação em Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - M do C P Nunes
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - K J Gollob
- Núcleo de Ensino e Pesquisa, Instituto Mário Pena, Belo Horizonte, MG, Brazil.,BRISA Diagnósticos, Belo Horizonte, MG, Brazil.,Instituto Nacional de Ciência e Tecnologia Doenças Tropicais - INCT-DT, Belo Horizonte, MG, Brazil
| | - W O Dutra
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,Pós-graduação em Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.,Instituto Nacional de Ciência e Tecnologia Doenças Tropicais - INCT-DT, Belo Horizonte, MG, Brazil
| |
Collapse
|
11
|
Ishengoma E, Agaba M. Evolution of toll-like receptors in the context of terrestrial ungulates and cetaceans diversification. BMC Evol Biol 2017; 17:54. [PMID: 28209121 PMCID: PMC5314619 DOI: 10.1186/s12862-017-0901-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 02/02/2017] [Indexed: 11/17/2022] Open
Abstract
Background Toll-like receptors (TLRs) are the frontline actors in the innate immune response to various pathogens and are expected to be targets of natural selection in species adapted to habitats with contrasting pathogen burdens. The recent publication of genome sequences of giraffe and okapi together afforded the opportunity to examine the evolution of selected TLRs in broad range of terrestrial ungulates and cetaceans during their complex habitat diversification. Through direct sequence comparisons and standard evolutionary approaches, the extent of nucleotide and protein sequence diversity in seven Toll-like receptors (TLR2, TLR3, TLR4, TLR5, TLR7, TLR9 and TLR10) between giraffe and closely related species was determined. In addition, comparison of the patterning of key TLR motifs and domains between giraffe and related species was performed. The quantification of selection pressure and divergence on TLRs among terrestrial ungulates and cetaceans was also performed. Results Sequence analysis shows that giraffe has 94–99% nucleotide identity with okapi and cattle for all TLRs analyzed. Variations in the number of Leucine-rich repeats were observed in some of TLRs between giraffe, okapi and cattle. Patterning of key TLR domains did not reveal any significant differences in the domain architecture among giraffe, okapi and cattle. Molecular evolutionary analysis for selection pressure identifies positive selection on key sites for all TLRs examined suggesting that pervasive evolutionary pressure has taken place during the evolution of terrestrial ungulates and cetaceans. Analysis of positively selected sites showed some site to be part of Leucine-rich motifs suggesting functional relevance in species-specific recognition of pathogen associated molecular patterns. Notably, clade analysis reveals significant selection divergence between terrestrial ungulates and cetaceans in viral sensing TLR3. Mapping of giraffe TLR3 key substitutions to the structure of the receptor indicates that at least one of giraffe altered sites coincides with TLR3 residue known to play a critical role in receptor signaling activity. Conclusion There is overall structural conservation in TLRs among giraffe, okapi and cattle indicating that the mechanism for innate immune response utilizing TLR pathways may not have changed very much during the evolution of these species. However, a broader phylogenetic analysis revealed signatures of adaptive evolution among terrestrial ungulates and cetaceans, including the observed selection divergence in TLR3. This suggests that long term ecological dynamics has led to species-specific innovation and functional variation in the mechanisms mediating innate immunity in terrestrial ungulates and cetaceans. Electronic supplementary material The online version of this article (doi:10.1186/s12862-017-0901-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edson Ishengoma
- School of Life Science and Biongineering, The Nelson Mandela African Institution of Sciences and Technology, P.O. Box 447, Arusha, Tanzania. .,Mkwawa University College of Education, University of Dar es Salaam, P.O. Box 2513, Iringa, Tanzania.
| | - Morris Agaba
- School of Life Science and Biongineering, The Nelson Mandela African Institution of Sciences and Technology, P.O. Box 447, Arusha, Tanzania.,Biosciences Eastern and Central Africa, International Livestock Research Institute, Nairobi, GPO00100, Kenya
| |
Collapse
|
12
|
Gil-Jaramillo N, Motta FN, Favali CBF, Bastos IMD, Santana JM. Dendritic Cells: A Double-Edged Sword in Immune Responses during Chagas Disease. Front Microbiol 2016; 7:1076. [PMID: 27471496 PMCID: PMC4943928 DOI: 10.3389/fmicb.2016.01076] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/27/2016] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) are the most important member of the antigen presenting cells group due to their ability to recognize antigen at the infection site and their high specialized antigen internalization capacity. These cells have central role in connecting the innate and adaptive immune responses against Trypanosoma cruzi, the causative agent of Chagas disease. These first line defense cells modulate host immune response depending on type, maturation level, cytokine milieu and DC receptor involved in the interactions with T. cruzi, influencing the development of the disease clinic forms. Here, we present a review of DCs-T. cruzi interactions both in human and murine models, pointing out the parasite ability to manipulate DCs activity for the purpose of evading innate immune response and assuring its own survival and persistence.
Collapse
Affiliation(s)
- Natalia Gil-Jaramillo
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| | - Flávia N. Motta
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
- Faculdade de Ceilândia, Universidade de BrasíliaBrasília, Brazil
| | - Cecília B. F. Favali
- Laboratório de Biologia do Gene, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| | - Izabela M. D. Bastos
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| | - Jaime M. Santana
- Laboratório de Interação Patógeno-Hospedeiro, Instituto de Biologia, Universidade de BrasíliaBrasília, Brazil
| |
Collapse
|
13
|
Silva-Neto MAC, Lopes AH, Atella GC. Here, There, and Everywhere: The Ubiquitous Distribution of the Immunosignaling Molecule Lysophosphatidylcholine and Its Role on Chagas Disease. Front Immunol 2016; 7:62. [PMID: 26925065 PMCID: PMC4759257 DOI: 10.3389/fimmu.2016.00062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/08/2016] [Indexed: 12/17/2022] Open
Abstract
Chagas disease is a severe illness, which can lead to death if the patients are not promptly treated. The disease is caused by the protozoan parasite Trypanosoma cruzi, which is mostly transmitted by a triatomine insect vector. There are 8-10 million people infected with T. cruzi in the world, but the transmission of such disease by bugs occurs only in the Americas, especially Latin America. Chronically infected patients will develop cardiac diseases (30%) and up digestive, neurological, or mixed disorders (10%). Lysophosphatidylcholine (LPC) is the major phospholipid component of oxidized low-density lipoproteins associated with atherosclerosis-related tissue damage. Insect-derived LPC powerfully attracts inflammatory cells to the site of the insect bite, enhances parasite invasion, and inhibits the production of nitric oxide by T. cruzi-stimulated macrophages. The recognition of the ubiquitous presence of LPC on the vector saliva, its production by the parasite itself and its presence both on patient plasma and its role on diverse host × parasite interaction systems lead us to compare its distribution in nature with the title of the famous Beatles song "Here, There and Everywhere" recorded exactly 50 years ago in 1966. Here, we review the major findings pointing out the role of such molecule as an immunosignaling modulator of Chagas disease transmission. Also, we believe that future investigation of the connection of this ubiquity and the immune role of such molecule may lead in the future to novel methods to control parasite transmission, infection, and pathogenesis.
Collapse
Affiliation(s)
- Mário Alberto C Silva-Neto
- Programa de Biologia Molecular e Biotecnologia, CCS, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brazil
| | - Angela H Lopes
- Centro de Ciências da Saúde, Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária - Ilha do Fundão , Rio de Janeiro , Brazil
| | - Georgia C Atella
- Programa de Biologia Molecular e Biotecnologia, CCS, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brazil
| |
Collapse
|
14
|
Vázquez A, Ruiz-Rosado JDD, Terrazas LI, Juárez I, Gomez-Garcia L, Calleja E, Camacho G, Chávez A, Romero M, Rodriguez T, Espinoza B, Rodriguez-Sosa M. Mouse macrophage galactose-type lectin (mMGL) is critical for host resistance against Trypanosoma cruzi infection. Int J Biol Sci 2014; 10:909-20. [PMID: 25170304 PMCID: PMC4147224 DOI: 10.7150/ijbs.9214] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 07/05/2014] [Indexed: 12/27/2022] Open
Abstract
The C-type lectin receptor mMGL is expressed exclusively by myeloid antigen presenting cells (APC) such as dendritic cells (DC) and macrophages (Mφ), and it mediates binding to glycoproteins carrying terminal galactose and α- or β-N-acetylgalactosamine (Gal/GalNAc) residues. Trypanosoma cruzi (T. cruzi) expresses large amounts of mucin (TcMUC)-like glycoproteins. Here, we show by lectin-blot that galactose moieties are also expressed on the surface of T. cruzi. Male mMGL knockout (-/-) and wild-type (WT) C57BL/6 mice were infected intraperitoneally with 104T. cruzi trypomastigotes (Queretaro strain). Following T. cruzi infection, mMGL-/- mice developed higher parasitemia and higher mortality rates compared with WT mice. Although hearts from T. cruzi-infected WT mice presented few amastigote nests, mMGL-/- mice displayed higher numbers of amastigote nests. Compared with WT, Mφ from mMGL-/- mice had low production of nitric oxide (NO), interleukin (IL)-12 and tumor necrosis factor (TNF)-α in response to soluble T. cruzi antigens (TcAg). Interestingly, upon in vitro T. cruzi infection, mMGL-/- Mφ expressed lower levels of MHC-II and TLR-4 and harbored higher numbers of parasites, even when mMGL-/- Mφ were previously primed with IFN-γ or LPS/IFN-γ. These data suggest that mMGL plays an important role during T. cruzi infection, is required for optimal Mφ activation, and may synergize with TLR-4-induced pathways to produce TNF-α, IL-1β and NO during the early phase of infection.
Collapse
Affiliation(s)
- Alicia Vázquez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Juan de Dios Ruiz-Rosado
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Luis I Terrazas
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Imelda Juárez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Lorena Gomez-Garcia
- 2. Department of Immunology, Instituto Nacional de Cardiología "Ignacio Chávez," México, D.F. 14080 México
| | - Elsa Calleja
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Griselda Camacho
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Ana Chávez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Miriam Romero
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Tonathiu Rodriguez
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| | - Bertha Espinoza
- 3. Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México. México, D.F. 04510 México
| | - Miriam Rodriguez-Sosa
- 1. Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México (UNAM), C. P. 54090, Estado de México, México
| |
Collapse
|
15
|
Role of the Apt1 protein in polysaccharide secretion by Cryptococcus neoformans. EUKARYOTIC CELL 2013; 13:715-26. [PMID: 24337112 DOI: 10.1128/ec.00273-13] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Flippases are key regulators of membrane asymmetry and secretory mechanisms. Vesicular polysaccharide secretion is essential for the pathogenic mechanisms of Cryptococcus neoformans. On the basis of the observations that flippases are required for polysaccharide secretion in plants and the putative Apt1 flippase is required for cryptococcal virulence, we analyzed the role of this enzyme in polysaccharide release by C. neoformans, using a previously characterized apt1Δ mutant. Mutant and wild-type (WT) cells shared important phenotypic characteristics, including capsule morphology and dimensions, glucuronoxylomannan (GXM) composition, molecular size, and serological properties. The apt1Δ mutant, however, produced extracellular vesicles (EVs) with a lower GXM content and different size distribution in comparison with those of WT cells. Our data also suggested a defective intracellular GXM synthesis in mutant cells, in addition to changes in the architecture of the Golgi apparatus. These findings were correlated with diminished GXM production during in vitro growth, macrophage infection, and lung colonization. This phenotype was associated with decreased survival of the mutant in the lungs of infected mice, reduced induction of interleukin-6 (IL-6) cytokine levels, and inefficacy in colonization of the brain. Taken together, our results indicate that the lack of APT1 caused defects in both GXM synthesis and vesicular export to the extracellular milieu by C. neoformans via processes that are apparently related to the pathogenic mechanisms used by this fungus during animal infection.
Collapse
|
16
|
Gazos-Lopes F, Mesquita RD, Silva-Cardoso L, Senna R, Silveira AB, Jablonka W, Cudischevitch CO, Carneiro AB, Machado EA, Lima LG, Monteiro RQ, Nussenzveig RH, Folly E, Romeiro A, Vanbeselaere J, Mendonça-Previato L, Previato JO, Valenzuela JG, Ribeiro JMC, Atella GC, Silva-Neto MAC. Glycoinositolphospholipids from Trypanosomatids subvert nitric oxide production in Rhodnius prolixus salivary glands. PLoS One 2012; 7:e47285. [PMID: 23077586 PMCID: PMC3471836 DOI: 10.1371/journal.pone.0047285] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 09/14/2012] [Indexed: 11/23/2022] Open
Abstract
Background Rhodnius prolixus is a blood-sucking bug vector of Trypanosoma cruzi and T. rangeli. T. cruzi is transmitted by vector feces deposited close to the wound produced by insect mouthparts, whereas T. rangeli invades salivary glands and is inoculated into the host skin. Bug saliva contains a set of nitric oxide-binding proteins, called nitrophorins, which deliver NO to host vessels and ensure vasodilation and blood feeding. NO is generated by nitric oxide synthases (NOS) present in the epithelium of bug salivary glands. Thus, T. rangeli is in close contact with NO while in the salivary glands. Methodology/Principal Findings Here we show by immunohistochemical, biochemical and molecular techniques that inositolphosphate-containing glycolipids from trypanosomatids downregulate NO synthesis in the salivary glands of R. prolixus. Injecting insects with T. rangeli-derived glycoinositolphospholipids (Tr GIPL) or T. cruzi-derived glycoinositolphospholipids (Tc GIPL) specifically decreased NO production. Salivary gland treatment with Tc GIPL blocks NO production without greatly affecting NOS mRNA levels. NOS protein is virtually absent from either Tr GIPL- or Tc GIPL-treated salivary glands. Evaluation of NO synthesis by using a fluorescent NO probe showed that T. rangeli-infected or Tc GIPL-treated glands do not show extensive labeling. The same effect is readily obtained by treatment of salivary glands with the classical protein tyrosine phosphatase (PTP) inhibitor, sodium orthovanadate (SO). This suggests that parasite GIPLs induce the inhibition of a salivary gland PTP. GIPLs specifically suppressed NO production and did not affect other anti-hemostatic properties of saliva, such as the anti-clotting and anti-platelet activities. Conclusions/Significance Taken together, these data suggest that trypanosomatids have overcome NO generation using their surface GIPLs. Therefore, these molecules ensure parasite survival and may ultimately enhance parasite transmission.
Collapse
Affiliation(s)
- Felipe Gazos-Lopes
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Rafael Dias Mesquita
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Lívia Silva-Cardoso
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Raquel Senna
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Alan Barbosa Silveira
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Willy Jablonka
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Cecília Oliveira Cudischevitch
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Alan Brito Carneiro
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Ednildo Alcantara Machado
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luize G. Lima
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Queiroz Monteiro
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Evelize Folly
- Universidade Federal Fluminense, Instituto de Biologia. Campus Valonguinho, Prédio do Instituto de Biologia, Departamento de Biologia Celular e Molecular, Centro, Niterói, Rio de Janeiro, Brasil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Alexandre Romeiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorick Vanbeselaere
- Université de Lille 1, Unité de Glycobiologie Structurale et Fonctionnelle, Villeneuve d’Ascq, France
| | - Lucia Mendonça-Previato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - José Osvaldo Previato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - José Marcos Chaves Ribeiro
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Georgia Correa Atella
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - Mário Alberto Cardoso Silva-Neto
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
17
|
Rasouli M, Keshavarz M, Kalani M, Moravej A, Kiany S, Badiee P. Toll-like receptor 4 (TLR4) polymorphisms in Iranian patients with visceral leishmaniasis. Mol Biol Rep 2012; 39:10795-802. [PMID: 23053976 DOI: 10.1007/s11033-012-1973-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 10/01/2012] [Indexed: 12/22/2022]
Abstract
The role of Toll-like receptor (TLR) 4 in visceral leishmaniasis (VL), a disease caused by an obligate intracellular protozoan parasites belonging to the genus Leishmania, has been shown in the recent leishmaniasis experimental studies. As genetic host factors play an important role in the susceptibility and/or resistance to VL, the association between TLR4 gene mutations [A896G and C1196T single nucleotide polymorphisms (SNPs)] and VL was investigated. Genotyping of A896G (Asp299Gly) and C1196T (Thr399Ile) SNPs was performed in the patients with VL (N = 122) and ethnically matched controls (N = 155) using polymerase chain reaction-restriction fragment length polymorphism method. When VL patients and the controls were compared, no statistically significant differences were observed in A896G and C1196T alleles and genotypes (P > 0.05). The TLR4 A896G and C1196T were in moderate linkage disequilibrium in the controls and patients (r (2) = 0.497, 0.548 and D' = 0.705, 0.808, respectively), and haplotypes reconstructed from these SNPs were not significantly different between the aforementioned study groups. In conclusion, based on the results, TLR4 gene polymorphisms at the positions 896 and 1196 cannot be regarded as the major contributors to VL susceptibility among the Iranian population.
Collapse
Affiliation(s)
- Manoochehr Rasouli
- Department of Immunology, Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, 71937-11351, Shiraz, Iran.
| | | | | | | | | | | |
Collapse
|
18
|
Andrade D, Serra R, Svensjö E, Lima APC, Ramos ES, Fortes FS, Morandini ACF, Morandi V, Soeiro MDN, Tanowitz HB, Scharfstein J. Trypanosoma cruzi invades host cells through the activation of endothelin and bradykinin receptors: a converging pathway leading to chagasic vasculopathy. Br J Pharmacol 2012; 165:1333-47. [PMID: 21797847 DOI: 10.1111/j.1476-5381.2011.01609.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Independent studies in experimental models of Trypanosoma cruzi appointed different roles for endothelin-1 (ET-1) and bradykinin (BK) in the immunopathogenesis of Chagas disease. Here, we addressed the hypothesis that pathogenic outcome is influenced by functional interplay between endothelin receptors (ET(A)R and ET(B)R) and bradykinin B(2) receptors (B(2)R). EXPERIMENTAL APPROACH Intravital microscopy was used to determine whether ETR/B(2)R drives the accumulation of rhodamine-labelled leucocytes in the hamster cheek pouch (HCP). Inflammatory oedema was measured in the infected BALB/c paw of mice. Parasite invasion was assessed in CHO over-expressing ETRs, mouse cardiomyocytes, endothelium (human umbilical vein endothelial cells) or smooth muscle cells (HSMCs), in the presence/absence of antagonists of B(2)R (HOE-140), ET(A)R (BQ-123) and ET(B)R (BQ-788), specific IgG antibodies to each GPCRs; cholesterol or calcium-depleting drugs. RNA interference (ET(A)R or ET(B)R genes) in parasite infectivity was investigated in HSMCs. KEY RESULTS BQ-123, BQ-788 and HOE-140 reduced leucocyte accumulation in HCP topically exposed to trypomastigotes and blocked inflammatory oedema in infected mice. Acting synergistically, ET(A)R and ET(B)R antagonists reduced parasite invasion of HSMCs to the same extent as HOE-140. Exogenous ET-1 potentiated T. cruzi uptake by HSMCs via ETRs/B(2)R, whereas RNA interference of ET(A)R and ET(B)R genes conversely reduced parasite internalization. ETRs/B(2)R-driven infection in HSMCs was reduced in HSMC pretreated with methyl-β-cyclodextrin, a cholesterol-depleting drug, or in thapsigargin- or verapamil-treated target cells. CONCLUSIONS AND IMPLICATIONS Our findings suggest that plasma leakage, a neutrophil-driven inflammatory response evoked by trypomastigotes via the kinin/endothelin pathways, may offer a window of opportunity for enhanced parasite invasion of cardiovascular cells.
Collapse
Affiliation(s)
- Daniele Andrade
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Rodrigues MM, Oliveira AC, Bellio M. The Immune Response to Trypanosoma cruzi: Role of Toll-Like Receptors and Perspectives for Vaccine Development. J Parasitol Res 2012; 2012:507874. [PMID: 22496959 PMCID: PMC3306967 DOI: 10.1155/2012/507874] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/31/2011] [Indexed: 02/06/2023] Open
Abstract
In the past ten years, studies have shown the recognition of Trypanosoma cruzi-associated molecular patterns by members of the Toll-like receptor (TLR) family and demonstrated the crucial participation of different TLRs during the experimental infection with this parasite. In the present review, we will focus on the role of TLR-activated pathways in the modulation of both innate and acquired immune responses to T. cruzi infection, as well as discuss the state of the art of vaccine research and development against the causative agent of Chagas disease (or American trypanosomiasis).
Collapse
Affiliation(s)
- Mauricio M. Rodrigues
- Centro de Terapia Celular e Molecular (CTCMol), Universidade Federal de São Paulo (UNIFESP), 04044-010 São Paulo, SP, Brazil
| | - Ana Carolina Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), 21941-902 Rio de Janeiro, RJ, Brazil
| | - Maria Bellio
- Instituto de Microbiologia Paulo de Góes, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro (UFRJ), CCS, Avenida Carlos Chagas Filho, 373 Bloco D, sala 35, Cidade Universitária, 21941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
20
|
Lysophosphatidylcholine: A Novel Modulator of Trypanosoma cruzi Transmission. J Parasitol Res 2011; 2012:625838. [PMID: 22132309 PMCID: PMC3206328 DOI: 10.1155/2012/625838] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 07/29/2011] [Accepted: 09/12/2011] [Indexed: 01/18/2023] Open
Abstract
Lysophosphatidylcholine is a bioactive lipid that regulates a large number of cellular processes and is especially present during the deposition and infiltration of inflammatory cells and deposition of atheromatous plaque. Such molecule is also present in saliva and feces of the hematophagous organism Rhodnius prolixus, a triatominae bug vector of Chagas disease. We have recently demonstrated that LPC is a modulator of Trypanosoma cruzi transmission. It acts as a powerful chemoattractant for inflammatory cells at the site of the insect bite, which will provide a concentrated population of cells available for parasite infection. Also, LPC increases macrophage intracellular calcium concentrations that ultimately enhance parasite invasion. Finally, LPC inhibits NO production by macrophages stimulated by live T. cruzi, and thus interferes with the immune system of the vertebrate host. In the present paper, we discuss the main signaling mechanisms that are likely used by such molecule and their eventual use as targets to block parasite transmission and the pathogenesis of Chagas disease.
Collapse
|
21
|
Dasgupta G, Chentoufi AA, You S, Falatoonzadeh P, Urbano LAA, Akhtarmalik A, Nguyen K, Ablabutyan L, Nesburn AB, BenMohamed L. Engagement of TLR2 reverses the suppressor function of conjunctiva CD4+CD25+ regulatory T cells and promotes herpes simplex virus epitope-specific CD4+CD25- effector T cell responses. Invest Ophthalmol Vis Sci 2011; 52:3321-33. [PMID: 21273544 DOI: 10.1167/iovs.10-6522] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE. The authors recently reported that Foxp3(+)CD4(+) CD25(+(Bright)) "natural" regulatory T cells (nT(reg) cells) are abundant in rabbit conjunctiva and suppress herpes simplex virus (HSV)-1-specific CD4(+) and CD8(+) effector T cells (T(eff) cells). However, little is known about the overall regulatory mechanisms of these nT(reg) cells. The authors investigate the regulation of conjunctiva-resident nT(reg) cells through Toll-like receptors (TLRs) and their effect on ocular mucosal T(eff) cell immunity. METHODS. CD4(+)CD25(+) nT(reg) cells were purified from naive rabbit conjunctivas, and their TLR expression profile was determined. The effects of TLR engagement on nT(reg) cell-mediated suppression of CD4(+) T(eff) cells were determined in vitro and in vivo. RESULTS. The authors found that conjunctiva-resident nT(reg) cells express high levels of TLR2 and TLR9; exposure to the TLR2 ligand lipoteichoic acid (LTA) led to the increased activation and proliferation of nT(reg) cells, and the addition of autologous APCs further increased nT(reg) cell expansion; in contrast, the TLR9 ligand CpG(2007) inhibited the proliferation of nT(reg) cells, and the addition of autologous APCs had no effect on such inhibition; nT(reg) cells treated with LTA, but not with CpG(2007), expressed IFN-γ and IL-10 mRNA, but not TGF-β; consistent with in vitro data, rabbits immunized by topical ocular drops of HSV-gD peptides + TLR2 ligand (LTA) displayed enhanced CD4(+)CD25(-) T(eff) cell immune responses when compared with HSV-gD peptides + TLR9 ligand (CpG(2007)). CONCLUSIONS. Although conjunctiva-resident CD4(+)CD25(+) nT(reg) cells express high level of TLR2 and TLR9, their suppressive function is more significantly reversed after the administration of TLR2 ligand (LTA; P < 0.005) than of TLR9 ligand (CpG(200); P > 0.005). These findings will likely help optimize the topical ocular administration of immunotherapies.
Collapse
Affiliation(s)
- Gargi Dasgupta
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California at Irvine, California
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Villani FNA, da Costa Rocha MO, Nunes MDCP, Antonelli LRDV, Magalhães LMD, dos Santos JSC, Gollob KJ, Dutra WO. Trypanosoma cruzi-induced activation of functionally distinct αβ and γδ CD4- CD8- T cells in individuals with polar forms of Chagas' disease. Infect Immun 2010; 78:4421-30. [PMID: 20696836 PMCID: PMC2950361 DOI: 10.1128/iai.00179-10] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Revised: 03/29/2010] [Accepted: 07/27/2010] [Indexed: 12/31/2022] Open
Abstract
CD4(-) CD8(-) (double-negative [DN]) T cells have recently been shown to display important immunological functions in human diseases. They express γδ or αβ T-cell receptors that recognize lipid/glycolipid antigens presented via the nonclassical major histocompatibility complex molecules of the CD1 family. We recently demonstrated that while αβ DN T cells serve primarily to express inflammatory cytokines, γδ DN T cells express mainly interleukin-10 (IL-10) in patients with cutaneous leishmaniasis. We also demonstrated a correlation between DN T cells and the expression of gamma interferon in the acute phase of Trypanosoma cruzi experimental infection. In this work, we sought to investigate whether αβ or γδ DN T cells display distinct immunoregulatory potentials in patients with polar forms of human Chagas' disease. Our data showed that in vitro infection with T. cruzi leads to expansion of DN T cells in patients with the indeterminate and severe cardiac clinical forms of the disease. However, while αβ DN T cells primarily produce inflammatory cytokines in both forms of the disease, γδ DN T cells display a marked, significant increase in antigen-specific IL-10 expression in indeterminate patients relative to cardiac patients. Finally, higher frequencies of the IL-10-producing γδ DN T cells were correlated with improved clinical measures of cardiac function in the patients, suggesting a protective role for these cells in Chagas' disease. Taken together, these data show distinct functional characteristics for αβ and γδ DN T cells associated with distinct morbidity rates and clinical forms in human Chagas' disease.
Collapse
Affiliation(s)
- Fernanda Nobre Amaral Villani
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| | - Manoel Otávio da Costa Rocha
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| | - Maria do Carmo Pereira Nunes
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| | - Lis Ribeiro do Valle Antonelli
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| | - Luisa Mourão Dias Magalhães
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| | - Janete Soares Coelho dos Santos
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| | - Kenneth J. Gollob
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| | - Walderez O. Dutra
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Infectious Diseases and Tropical Medicine Graduate Course, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil, Instituto Oswald Cruz, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil, SRI International, Biosciences Division, Center for Infectious Disease Research, Menlo Park, California, Graduate Program in Biosciences and Medicine, Santa Casa Hospital, Belo Horizonte, Minas Gerais, Brazil, INCT-DT, Brazil
| |
Collapse
|
23
|
Benítez-Hernández I, Méndez-Enríquez E, Ostoa P, Fortoul T, Ramírez J, Stempin C, Cerbán F, Soldevila G, García-Zepeda E. Proteolytic cleavage of chemokines by Trypanosoma cruzi's cruzipain inhibits chemokine functions by promoting the generation of antagonists. Immunobiology 2010; 215:413-26. [DOI: 10.1016/j.imbio.2009.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Revised: 06/04/2009] [Accepted: 06/08/2009] [Indexed: 01/04/2023]
|
24
|
Oliveira AC, de Alencar BC, Tzelepis F, Klezewsky W, da Silva RN, Neves FS, Cavalcanti GS, Boscardin S, Nunes MP, Santiago MF, Nóbrega A, Rodrigues MM, Bellio M. Impaired innate immunity in Tlr4(-/-) mice but preserved CD8+ T cell responses against Trypanosoma cruzi in Tlr4-, Tlr2-, Tlr9- or Myd88-deficient mice. PLoS Pathog 2010; 6:e1000870. [PMID: 20442858 PMCID: PMC2861687 DOI: 10.1371/journal.ppat.1000870] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Accepted: 03/23/2010] [Indexed: 01/12/2023] Open
Abstract
The murine model of T. cruzi infection has provided compelling evidence that development of host resistance against intracellular protozoans critically depends on the activation of members of the Toll-like receptor (TLR) family via the MyD88 adaptor molecule. However, the possibility that TLR/MyD88 signaling pathways also control the induction of immunoprotective CD8+ T cell-mediated effector functions has not been investigated to date. We addressed this question by measuring the frequencies of IFN-γ secreting CD8+ T cells specific for H-2Kb-restricted immunodominant peptides as well as the in vivo Ag-specific cytotoxic response in infected animals that are deficient either in TLR2, TLR4, TLR9 or MyD88 signaling pathways. Strikingly, we found that T. cruzi-infected Tlr2−/−, Tlr4−/−, Tlr9−/− or Myd88−/− mice generated both specific cytotoxic responses and IFN-γ secreting CD8+ T cells at levels comparable to WT mice, although the frequency of IFN-γ+CD4+ cells was diminished in infected Myd88−/− mice. We also analyzed the efficiency of TLR4-driven immune responses against T. cruzi using TLR4-deficient mice on the C57BL genetic background (B6 and B10). Our studies demonstrated that TLR4 signaling is required for optimal production of IFN-γ, TNF-α and nitric oxide (NO) in the spleen of infected animals and, as a consequence, Tlr4−/− mice display higher parasitemia levels. Collectively, our results indicate that TLR4, as well as previously shown for TLR2, TLR9 and MyD88, contributes to the innate immune response and, consequently, resistance in the acute phase of infection, although each of these pathways is not individually essential for the generation of class I-restricted responses against T. cruzi. Innate and acquired immune responses are triggered during infection with T. cruzi, the etiologic agent of Chagas' disease, and are critical for host survival. Parasite burden is usually controlled by the time the adaptive response becomes operational. Nevertheless, T. cruzi manages to subsist within intracellular niches and establishes a chronic infection, leading to the development of cardiomyopathy in approximately one-third of infected individuals. Recently, Toll-like receptors (TLRs) have been shown to recognize T. cruzi molecules and mice lacking MyD88, the key adaptor for most TLRs, are extremely susceptible to infection. Although TLRs are known to link innate and adaptive responses, their role in the establishment of crucial effector mechanisms mediated by CD8+ T cells during T. cruzi infection has not yet been determined. We analyzed the induction of IFN-γ and cytotoxic activity in vivo in TLR2-, TLR4-, TLR9- or MyD88-deficient mice during infection, and found intact responses compared to WT mice. We also demonstrated that TLR4 is required for optimal production of inflammatory cytokines and nitric oxide and, consequently, for a better control of parasitemia levels. Understanding how TLR activation leads to resistance to infection might contribute to the development of better strategies to improve immune responses against this pathogen.
Collapse
Affiliation(s)
- Ana-Carolina Oliveira
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruna C. de Alencar
- Centro Interdisciplinar de Terapia Gênica (CINTERGEN), Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Fanny Tzelepis
- Centro Interdisciplinar de Terapia Gênica (CINTERGEN), Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Weberton Klezewsky
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel N. da Silva
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabieni S. Neves
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gisele S. Cavalcanti
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silvia Boscardin
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Marise P. Nunes
- Instituto Osvaldo Cruz (IOC/FIOCRUZ) Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo F. Santiago
- Instituto de Biofísica Carlos Chagas Filho (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Nóbrega
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maurício M. Rodrigues
- Centro Interdisciplinar de Terapia Gênica (CINTERGEN), Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Maria Bellio
- Instituto de Microbiologia Prof. Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
25
|
Guedes PMDM, Gutierrez FRS, Maia FL, Milanezi CM, Silva GK, Pavanelli WR, Silva JS. IL-17 produced during Trypanosoma cruzi infection plays a central role in regulating parasite-induced myocarditis. PLoS Negl Trop Dis 2010; 4:e604. [PMID: 20169058 PMCID: PMC2821906 DOI: 10.1371/journal.pntd.0000604] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 12/22/2009] [Indexed: 01/12/2023] Open
Abstract
Background Chagas disease is a neglected disease caused by the intracellular parasite Trypanosoma cruzi. Around 30% of the infected patients develop chronic cardiomyopathy or megasyndromes, which are high-cost morbid conditions. Immune response against myocardial self-antigens and exacerbated Th1 cytokine production has been associated with the pathogenesis of the disease. As IL-17 is involved in the pathogenesis of several autoimmune, inflammatory and infectious diseases, we investigated its role during the infection with T. cruzi. Methodology/Principal Findings First, we detected significant amounts of CD4, CD8 and NK cells producing IL-17 after incubating live parasites with spleen cells from normal BALB/c mice. IL-17 is also produced in vivo by CD4+, CD8+ and NK cells from BALB/c mice on the early acute phase of infection. Treatment of infected mice with anti-mouse IL-17 mAb resulted in increased myocarditis, premature mortality, and decreased parasite load in the heart. IL-17 neutralization resulted in increased production of IL-12, IFN-γ and TNF-α and enhanced specific type 1 chemokine and chemokine receptors expression. Moreover, the results showed that IL-17 regulates T-bet, RORγt and STAT-3 expression in the heart, showing that IL-17 controls the differentiation of Th1 cells in infected mice. Conclusion/Significance These results show that IL-17 controls the resistance to T. cruzi infection in mice regulating the Th1 cells differentiation, cytokine and chemokine production and control parasite-induced myocarditis, regulating the influx of inflammatory cells to the heart tissue. Correlations between the levels of IL-17, the extent of myocardial destruction, and the evolution of cardiac disease could identify a clinical marker of disease progression and may help in the design of alternative therapies for the control of chronic morbidity of chagasic patients. Chagas disease is caused by the intracellular parasite Trypanosoma cruzi. This infection has been considered one of the most neglected diseases and affects several million people in the Central and South America. Around 30% of the infected patients develop digestive and cardiac forms of the disease. Most patients are diagnosed during the chronic phase, when the treatment is not effective. Here, we showed by the first time that IL-17 is produced during experimental T. cruzi infection and that it plays a significant role in host defense, modulating parasite-induced myocarditis. Applying this analysis to humans could be of great value in unraveling the elements involved in the pathogenesis of chagasic cardiopathy and could be used in the development of alternative therapies to reduce morbidity during the chronic phase of the disease, as well as clinical markers of disease progression. The understanding of these aspects of disease may be helpful in reducing the disability-adjusted life years (DALYs) and costs to the public health service in developing countries.
Collapse
Affiliation(s)
- Paulo Marcos da Matta Guedes
- Department of Biochemistry and immunology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fredy R. S. Gutierrez
- Department of Biochemistry and immunology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Flavia L. Maia
- Department of Biochemistry and immunology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Cristiane M. Milanezi
- Department of Biochemistry and immunology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Grace K. Silva
- Department of Biochemistry and immunology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Wander R. Pavanelli
- Department of Biochemistry and immunology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - João S. Silva
- Department of Biochemistry and immunology, School of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
26
|
Innate immune sensing and activation of cell surface Toll-like receptors. Semin Immunol 2009; 21:175-84. [DOI: 10.1016/j.smim.2009.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Accepted: 05/06/2009] [Indexed: 12/30/2022]
|
27
|
Scharfstein J, Gomes JDAS, Correa-Oliveira R. Back to the future in Chagas disease: from animal models to patient cohort studies, progress in immunopathogenesis research. Mem Inst Oswaldo Cruz 2009; 104 Suppl 1:187-98. [DOI: 10.1590/s0074-02762009000900025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Accepted: 06/17/2009] [Indexed: 11/22/2022] Open
|
28
|
|