1
|
Damasceno TR, Tanaka DM, Magnani EF, Oliveira RDB, Pereira DAG, Vieira-Alves I, Lemos VS, Cabeza JM, Fabricio CG, Resende AA, Gonçalves DAP, Zanetti GDO, de Carvalho EEV, Simões MV, Oliveira LFL. Exercise Training Reduces Inflammation and Fibrosis and Preserves Myocardial Function and Perfusion in a Model of Chronic Chagas Cardiomyopathy. Arq Bras Cardiol 2024; 121:e20230707. [PMID: 39258653 PMCID: PMC11495816 DOI: 10.36660/abc.20230707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/05/2024] [Accepted: 04/24/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Chronic Chagas cardiomyopathy (CCC) is caused by an inflammatory process induced by Trypanosoma cruzi, which leads to myocarditis with reactive and reparative fibrosis. CCC progresses with myocardial perfusion abnormalities and histopathological events that affect cardiorespiratory fitness (CRF). OBJECTIVES We evaluated the effects of aerobic physical training (APT) on myocardial perfusion and on morphological and functional impairments related with inflammation and fibrosis in Syrian hamsters with CCC. As a secondary objective, we analyzed the cross-sectional areas of the skeletal muscle. METHODS Hamsters with CCC and their respective controls were divided into four groups: CCC sedentary, CCC-APT, sedentary control and APT control. Seven months after infection, the animals underwent echocardiography, myocardial perfusion scintigraphy and cardiopulmonary exercise testing. Moderate-intensity APT was performed for fifty minutes, five times a week, for eight weeks. Subsequently, the animals were reassessed. Histopathological analysis was conducted after the above-mentioned procedures. The level of significance was set at 5% in all analyses (p<0.05). RESULTS CCC sedentary animals presented worse myocardial perfusion defects (MPD) over time, reduced left ventricle ejection fraction (LVEF) and showed more inflammation and fibrosis when compared to other groups (mixed ANOVA analysis). Conversely, APT was able to mitigate the progression of MPD, ameliorate inflammation and fibrosis and improve CRF efficiency in CCC-APT animals. CONCLUSIONS Our study demonstrated that APT ameliorated cardiac dysfunction, MPD, and reduced inflammation and fibrosis in CCC hamster models. Additionally, CCC-SED animals presented skeletal muscle atrophy while CCC-APT animals showed preserved skeletal muscle CSA. Understanding APT's effects on CCC's pathophysiological dimensions is crucial for future research and therapeutic interventions.
Collapse
Affiliation(s)
- Thayrine R. Damasceno
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Denise M. Tanaka
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Enrico F. Magnani
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Rafael D. B. Oliveira
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Danielle A. G. Pereira
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Ildernandes Vieira-Alves
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Virginia S. Lemos
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Jorge M. Cabeza
- Hospital Israelita Albert EinsteinSão PauloSPBrasilHospital Israelita Albert Einstein, São Paulo, SP – Brasil
| | - Camila G. Fabricio
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Alessandra A. Resende
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Dawit A. P. Gonçalves
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Gustavo de Oliveira Zanetti
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
| | - Eduardo E. Vieira de Carvalho
- Universidade Federal do Triângulo MineiroUberabaMGBrasilUniversidade Federal do Triângulo Mineiro, Uberaba, MG – Brasil
| | - Marcus V. Simões
- Universidade de São PauloFaculdade de Medicina de Ribeirão PretoRibeirão PretoSPBrasilFaculdade de Medicina de Ribeirão Preto – Universidade de São Paulo, Ribeirão Preto, SP – Brasil
| | - Luciano F. L. Oliveira
- Universidade Federal de Minas GeraisBelo HorizonteMGBrasilUniversidade Federal de Minas Gerais, Belo Horizonte, MG – Brasil
- Universidade Federal do Triângulo MineiroUberabaMGBrasilUniversidade Federal do Triângulo Mineiro, Uberaba, MG – Brasil
| |
Collapse
|
2
|
Ambrosio LF, Volpini X, Quiroz JN, Brugo MB, Knubel CP, Herrera MR, Fozzatti L, Avila Pacheco J, Clish CB, Takenaka MC, Beloscar J, Theumer MG, Quintana FJ, Perez AR, Motrán CC. Association between altered tryptophan metabolism, plasma aryl hydrocarbon receptor agonists, and inflammatory Chagas disease. Front Immunol 2024; 14:1267641. [PMID: 38283348 PMCID: PMC10811785 DOI: 10.3389/fimmu.2023.1267641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/26/2023] [Indexed: 01/30/2024] Open
Abstract
Introduction Chagas disease causes a cardiac illness characterized by immunoinflammatory reactions leading to myocardial fibrosis and remodeling. The development of Chronic Chagas Cardiomyopathy (CCC) in some patients while others remain asymptomatic is not fully understood, but dysregulated inflammatory responses are implicated. The Aryl hydrocarbon receptor (AhR) plays a crucial role in regulating inflammation. Certain tryptophan (Trp) metabolites have been identified as AhR ligands with regulatory functions. Methods results and discussion We investigated AhR expression, agonist response, ligand production, and AhR-dependent responses, such as IDO activation and regulatory T (Treg) cells induction, in two T. cruzi-infected mouse strains (B6 and Balb/c) showing different polymorphisms in AhR. Furthermore, we assessed the metabolic profile of Trp catabolites and AhR agonistic activity levels in plasma samples from patients with chronic Chagas disease (CCD) and healthy donors (HD) using a luciferase reporter assay and liquid chromatography-mass spectrophotometry (LC-MS) analysis. T. cruzi-infected B6 mice showed impaired AhR-dependent responses compared to Balb/c mice, including reduced IDO activity, kynurenine levels, Treg cell induction, CYP1A1 up-regulation, and AhR expression following agonist activation. Additionally, B6 mice exhibited no detectable AhR agonist activity in plasma and displayed lower CYP1A1 up-regulation and AhR expression upon agonist activation. Similarly, CCC patients had decreased AhR agonistic activity in plasma compared to HD patients and exhibited dysregulation in Trp metabolic pathways, resulting in altered plasma metabolite profiles. Notably, patients with severe CCC specifically showed increased N-acetylserotonin levels in their plasma. The methods and findings presented here contribute to a better understanding of CCC development mechanisms and may identify potential specific biomarkers for T. cruzi infection and the severity of associated heart disease. These insights could be valuable in designing new therapeutic strategies. Ultimately, this research aims to establish the AhR agonistic activity and Trp metabolic profile in plasma as an innovative, non-invasive predictor of prognosis for chronic Chagas disease.
Collapse
Affiliation(s)
- Laura Fernanda Ambrosio
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Ximena Volpini
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Juan Nahuel Quiroz
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - María Belén Brugo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Carolina Paola Knubel
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Melisa Rocío Herrera
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Fozzatti
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Julián Avila Pacheco
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Clary B. Clish
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Maisa C. Takenaka
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Juan Beloscar
- Servicio de Cardiología, Departamento de Chagas, Hospital Provincial del Centenario y Cátedra de Cardiología, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Martín Gustavo Theumer
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Francisco Javier Quintana
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Ana Rosa Perez
- Instituto de Inmunología Clínica y Experimental de Rosario-CONICET-Universidad Nacional de Rosario (IDICER-CONICET-UNR), Rosario, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| |
Collapse
|
3
|
Jones KM, Zhan B, Ernste KJ, Villar MJ, Bisht N, Nguyen D, Chang LY, Poveda C, Robinson GJ, Trivedi AJ, Hofferek CJ, Decker WK, Konduri V. Immunomodulatory proteins from hookworms reduce cardiac inflammation and modulate regulatory responses in a mouse model of chronic Trypanosoma cruzi infection. FRONTIERS IN PARASITOLOGY 2023; 2:1244604. [PMID: 38239430 PMCID: PMC10795693 DOI: 10.3389/fpara.2023.1244604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
Introduction Hookworms are parasitic helminths that secrete a variety of proteins that induce anti-inflammatory immune responses, stimulating increased CD4 + Foxp3+ regulatory T cells and IL-10 production. Hookworm-derived recombinant proteins AIP-1 and AIP-2 have been shown to reduce inflammation in mouse models of inflammatory bowel disease and inflammatory airway disease by inducing CD4+Foxp3+ cells and IL-10 production. In contrast, chronic infection with the protozoal parasite Trypanosoma cruzi, the causative agent of Chagas disease, leads to chronic inflammation in tissues. Persistence of the parasites in tissues drives chronic low-grade inflammation, with increased infiltration of inflammatory cells into the heart, accompanied by increased production of inflammatory cytokines. There are no current antiparasitic drugs that effectively reduce or prevent chronic myocarditis caused by the onset of Chagas disease, thus new therapies are urgently needed. Therefore, the impact of AIP-1 and AIP-2 on myocarditis was investigated in a mouse model of chronic T. cruzi infection. Methods Female BALB/c mice infected with bioluminescent T. cruzi H1 strain trypomastigotes for 70 days were treated once daily for 7 days with 1mg/kg AIP-1 or AIP-2 protein by intraperitoneal injection. Control mice were left untreated or treated once daily for 14 days with 25mg/kg aspirin in drinking water. At 84 days of infection, splenocytes, cardiac tissue and serum were collected for evaluation. Results Treatment with both AIP-1 and AIP-2 proteins significantly reduced cardiac cellular infiltration, and reduced cardiac levels of IFNγ, IL-6 and IL-2. AIP-2 treatment reduced cardiac expression of COX-2. Further, while incubation with AIP-1 and AIP-2 proteins did not induce a significant upregulation of an immunoregulatory phenotype in dendritic cells (DC), there was a modest upregulation of CD11c +CD11b+MHCII+SIRPα+ expression, suggesting a regulatory phenotype. Ex-vivo stimulation of splenocytes from the treatment groups with AIP-1 loaded DC induced reduced levels of cytotoxic and pro-inflammatory T cells, stimulation with AIP-2 loaded DC specifically induced enhanced levels of CD4+CD25+Foxp3+ regulatory T cells among treatment groups. Discussion All in vivo and in vitro results demonstrate that hookworm-derived AIP-1 and AIP-2 proteins reduce T. cruzi induced cardiac inflammation, possibly through multiple anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Kathryn M. Jones
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Bin Zhan
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Keenan J. Ernste
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Maria Jose Villar
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Nalini Bisht
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Duc Nguyen
- Center for Comparative Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Li-Yen Chang
- Department of Medical Microbiology, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Cristina Poveda
- National School of Tropical Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX, United States
| | - Gonteria J. Robinson
- Molecular & Human Genetics Department, Baylor College of Medicine, Houston, TX, United States
| | - Akshar J. Trivedi
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Colby J. Hofferek
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - William K. Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer, Baylor College of Medicine, Houston, TX, United States
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, United States
| | - Vanaja Konduri
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
- Dan L. Duncan Cancer, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
4
|
Marin-Neto JA, Rassi A, Oliveira GMM, Correia LCL, Ramos Júnior AN, Luquetti AO, Hasslocher-Moreno AM, Sousa ASD, Paola AAVD, Sousa ACS, Ribeiro ALP, Correia Filho D, Souza DDSMD, Cunha-Neto E, Ramires FJA, Bacal F, Nunes MDCP, Martinelli Filho M, Scanavacca MI, Saraiva RM, Oliveira Júnior WAD, Lorga-Filho AM, Guimarães ADJBDA, Braga ALL, Oliveira ASD, Sarabanda AVL, Pinto AYDN, Carmo AALD, Schmidt A, Costa ARD, Ianni BM, Markman Filho B, Rochitte CE, Macêdo CT, Mady C, Chevillard C, Virgens CMBD, Castro CND, Britto CFDPDC, Pisani C, Rassi DDC, Sobral Filho DC, Almeida DRD, Bocchi EA, Mesquita ET, Mendes FDSNS, Gondim FTP, Silva GMSD, Peixoto GDL, Lima GGD, Veloso HH, Moreira HT, Lopes HB, Pinto IMF, Ferreira JMBB, Nunes JPS, Barreto-Filho JAS, Saraiva JFK, Lannes-Vieira J, Oliveira JLM, Armaganijan LV, Martins LC, Sangenis LHC, Barbosa MPT, Almeida-Santos MA, Simões MV, Yasuda MAS, Moreira MDCV, Higuchi MDL, Monteiro MRDCC, Mediano MFF, Lima MM, Oliveira MTD, Romano MMD, Araujo NNSLD, Medeiros PDTJ, Alves RV, Teixeira RA, Pedrosa RC, Aras Junior R, Torres RM, Povoa RMDS, Rassi SG, Alves SMM, Tavares SBDN, Palmeira SL, Silva Júnior TLD, Rodrigues TDR, Madrini Junior V, Brant VMDC, Dutra WO, Dias JCP. SBC Guideline on the Diagnosis and Treatment of Patients with Cardiomyopathy of Chagas Disease - 2023. Arq Bras Cardiol 2023; 120:e20230269. [PMID: 37377258 PMCID: PMC10344417 DOI: 10.36660/abc.20230269] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023] Open
Affiliation(s)
- José Antonio Marin-Neto
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Anis Rassi
- Hospital do Coração Anis Rassi , Goiânia , GO - Brasil
| | | | | | | | - Alejandro Ostermayer Luquetti
- Centro de Estudos da Doença de Chagas , Hospital das Clínicas da Universidade Federal de Goiás , Goiânia , GO - Brasil
| | | | - Andréa Silvestre de Sousa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Antônio Carlos Sobral Sousa
- Universidade Federal de Sergipe , São Cristóvão , SE - Brasil
- Hospital São Lucas , Rede D`Or São Luiz , Aracaju , SE - Brasil
| | | | | | | | - Edecio Cunha-Neto
- Universidade de São Paulo , Faculdade de Medicina da Universidade, São Paulo , SP - Brasil
| | - Felix Jose Alvarez Ramires
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Fernando Bacal
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Martino Martinelli Filho
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Maurício Ibrahim Scanavacca
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Magalhães Saraiva
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Adalberto Menezes Lorga-Filho
- Instituto de Moléstias Cardiovasculares , São José do Rio Preto , SP - Brasil
- Hospital de Base de Rio Preto , São José do Rio Preto , SP - Brasil
| | | | | | - Adriana Sarmento de Oliveira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Ana Yecê das Neves Pinto
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | | | - Andre Schmidt
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | - Andréa Rodrigues da Costa
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Barbara Maria Ianni
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Carlos Eduardo Rochitte
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Hcor , Associação Beneficente Síria , São Paulo , SP - Brasil
| | | | - Charles Mady
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Christophe Chevillard
- Institut National de la Santé Et de la Recherche Médicale (INSERM), Marselha - França
| | | | | | | | - Cristiano Pisani
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | - Edimar Alcides Bocchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Evandro Tinoco Mesquita
- Hospital Universitário Antônio Pedro da Faculdade Federal Fluminense , Niterói , RJ - Brasil
| | | | | | | | | | | | - Henrique Horta Veloso
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| | - Henrique Turin Moreira
- Hospital das Clínicas , Faculdade de Medicina de Ribeirão Preto , Universidade de São Paulo , Ribeirão Preto , SP - Brasil
| | | | | | | | - João Paulo Silva Nunes
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
- Fundação Zerbini, Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | | | | | | | - Luiz Cláudio Martins
- Universidade Estadual de Campinas , Faculdade de Ciências Médicas , Campinas , SP - Brasil
| | | | | | | | - Marcos Vinicius Simões
- Universidade de São Paulo , Faculdade de Medicina de Ribeirão Preto , Ribeirão Preto , SP - Brasil
| | | | | | - Maria de Lourdes Higuchi
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | - Mauro Felippe Felix Mediano
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
- Instituto Nacional de Cardiologia (INC), Rio de Janeiro, RJ - Brasil
| | - Mayara Maia Lima
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | | | | | - Renato Vieira Alves
- Instituto René Rachou , Fundação Oswaldo Cruz , Belo Horizonte , MG - Brasil
| | - Ricardo Alkmim Teixeira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | - Roberto Coury Pedrosa
- Hospital Universitário Clementino Fraga Filho , Instituto do Coração Edson Saad - Universidade Federal do Rio de Janeiro , RJ - Brasil
| | | | | | | | | | - Silvia Marinho Martins Alves
- Ambulatório de Doença de Chagas e Insuficiência Cardíaca do Pronto Socorro Cardiológico Universitário da Universidade de Pernambuco (PROCAPE/UPE), Recife , PE - Brasil
| | | | - Swamy Lima Palmeira
- Secretaria de Vigilância em Saúde , Ministério da Saúde , Brasília , DF - Brasil
| | | | | | - Vagner Madrini Junior
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo , São Paulo , SP - Brasil
| | | | | | - João Carlos Pinto Dias
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz , Rio de Janeiro , RJ - Brasil
| |
Collapse
|
5
|
Jones KM, Mangin EN, Reynolds CL, Villanueva LE, Cruz JV, Versteeg L, Keegan B, Kendricks A, Pollet J, Gusovsky F, Bottazzi ME, Hotez PJ. Vaccine-linked chemotherapy improves cardiac structure and function in a mouse model of chronic Chagas disease. Front Cell Infect Microbiol 2023; 13:1106315. [PMID: 36844399 PMCID: PMC9947347 DOI: 10.3389/fcimb.2023.1106315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Chagas disease, caused by chronic infection with the protozoan parasite Trypanosoma cruzi, affects 6-7 million people worldwide. The major clinical manifestation of Chagas disease is chronic Chagasic cardiomyopathy (CCC), which encompasses a spectrum of symptoms including arrhythmias, hypertrophy, dilated cardiomyopathy, heart failure, and sudden death. Current treatment is limited to two antiparasitic drugs, benznidazole (BNZ) and nifurtimox, but both have limited efficacy to halt the progression of CCC. We developed a vaccine-linked chemotherapy strategy using our vaccine consisting of recombinant Tc24-C4 protein and a TLR-4 agonist adjuvant in a stable squalene emulsion, in combination with low dose benznidazole treatment. We previously demonstrated in acute infection models that this strategy parasite specific immune responses, and reduced parasite burdens and cardiac pathology. Here, we tested our vaccine-linked chemotherapy strategy in a mouse model of chronic T. cruzi infection to evaluate the effect on cardiac function. Methods Female BALB/c mice infected with 500 blood form T. cruzi H1 strain trypomastigotes were treated beginning 70 days after infection with a low dose of BNZ and either low or high dose of vaccine, in both sequential and concurrent treatments streams. Control mice were untreated, or administered only one treatment. Cardiac health was monitored throughout the course of treatment by echocardiography and electrocardiograms. Approximately 8 months after infection, endpoint histopathology was performed to measure cardiac fibrosis and cellular infiltration. Results Vaccine-linked chemotherapy improved cardiac function as evidenced by amelioration of altered left ventricular wall thickness, left ventricular diameter, as well as ejection fraction and fractional shortening by approximately 4 months of infection, corresponding to two months after treatment was initiated. At study endpoint, vaccine-linked chemotherapy reduced cardiac cellular infiltration, and induced significantly increased antigen specific IFN-γ and IL-10 release from splenocytes, as well as a trend toward increased IL-17A. Discussion These data suggest that vaccine-linked chemotherapy ameliorates changes in cardiac structure and function induced by infection with T. cruzi. Importantly, similar to our acute model, the vaccine-linked chemotherapy strategy induced durable antigen specific immune responses, suggesting the potential for a long lasting protective effect. Future studies will evaluate additional treatments that can further improve cardiac function during chronic infection.
Collapse
Affiliation(s)
- Kathryn M. Jones
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States,*Correspondence: Kathryn M. Jones,
| | - Elise N. Mangin
- Department of Molecular Physiology, Baylor College of Medicine, Houston, TX, United States
| | - Corey L. Reynolds
- Department of Molecular Physiology, Baylor College of Medicine, Houston, TX, United States
| | - Liliana E. Villanueva
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Julio Vladimir Cruz
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States,Laboratorio de Parasitología, Centro de Investigaciones Regionales Dr. Hideyo Noguchi, Universidad Autónoma de Yucatán, Mérida, Yucatán, Mexico
| | - Leroy Versteeg
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States,Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, Netherlands
| | - Brian Keegan
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - April Kendricks
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Fabian Gusovsky
- Global Health Research, Eisai, Inc., Cambridge, MA, United States
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States,Department of Biology, Baylor University, Waco, TX, United States
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States,Department of Biology, Baylor University, Waco, TX, United States,James A. Baker III Institute for Public Policy, Rice University, Houston, TX, United States,Hagler Institute for Advanced Study at Texas A&M University, College Station, TX, United States
| |
Collapse
|
6
|
Khan R, Mirza MA, Aqil M, Alex TS, Raj N, Manzoor N, Naseef PP, Saheer Kuruniyan M, Iqbal Z. In Vitro and In Vivo Investigation of a Dual-Targeted Nanoemulsion Gel for the Amelioration of Psoriasis. Gels 2023; 9:gels9020112. [PMID: 36826282 PMCID: PMC9957534 DOI: 10.3390/gels9020112] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Psoriasis, due to its unique pathological manifestations and the limited success of existing therapeutic modalities, demands dedicated domain research. Our group has developed nanotherapeutics consisting of bioactives such as Thymoquinone (TQ) and Fulvic acid (FA), which have been successfully incorporated into a Nanoemulsion gel (NEG), taking kalonji oil as oil phase. The composition is aimed at ameliorating psoriasis with better therapeutic outcomes. TQ is a natural bio-active that has been linked to anti-psoriatic actions. FA has anti-inflammatory actions due to its free radical and oxidant-scavenging activity. Our previous publication reports the formulation development of the NEG, where we overcame the pharmaco-technical limitations of combining the above two natural bioactives. In vitro evaluation of the optimized NEG was carried out, which showed an enhanced dissolution rate and skin permeation of TQ. This work furthers the pharmaceutical progression of dual-targeted synergistic NEG to treat psoriasis. A suitable animal model, BALB/c mice, has been used to conduct the in vivo studies, which revealed the effective anti-psoriatic action of TQ. Molecular docking studies corroborated the results and revealed a good binding affinity for both the targets of TNF-α (Tumor necrosis factor) and IL-6 (Interlukin-6). Tissue uptake by Confocal laser scanning microscopy (CLSM), a skin interaction study of the gel formulation, and an antioxidant free radical scavenging assay (1-1 Diphenyl-2-picrylhydrazyl DPPH) were also carried out. It was concluded that the NEG may be effective in treating psoriasis with minimal side effects.
Collapse
Affiliation(s)
- Rahmuddin Khan
- Department of Pharmaceutics, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Mohd. Aamir Mirza
- Department of Pharmaceutics, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
- Correspondence: (M.A.M.); (Z.I.); Tel.: +91-9213378765 (M.A.M.); +91-9811733016 (Z.I.)
| | - Mohd Aqil
- Department of Pharmaceutics, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Thomson Santosh Alex
- Department of Pharmaceutics, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
| | - Nafis Raj
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Nikhat Manzoor
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | | | - Mohamed Saheer Kuruniyan
- Department of Dental Technology, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Zeenat Iqbal
- Department of Pharmaceutics, School of Pharmaceutical Education & Research (SPER), Jamia Hamdard, New Delhi 110062, India
- Correspondence: (M.A.M.); (Z.I.); Tel.: +91-9213378765 (M.A.M.); +91-9811733016 (Z.I.)
| |
Collapse
|
7
|
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
|
8
|
Jones KM, Poveda C, Versteeg L, Bottazzi ME, Hotez PJ. Preclinical advances and the immunophysiology of a new therapeutic chagas disease vaccine. Expert Rev Vaccines 2022; 21:1185-1203. [PMID: 35735065 DOI: 10.1080/14760584.2022.2093721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic infection with the protozoal parasite Trypanosoma cruzi leads to a progressive cardiac disease, known as chronic Chagasic cardiomyopathy (CCC). A new therapeutic Chagas disease vaccine is in development to augment existing antiparasitic chemotherapy drugs. AREAS COVERED We report on our current understanding of the underlying immunologic and physiologic mechanisms that lead to CCC, including parasite immune escape mechanisms that allow persistence and the subsequent inflammatory and fibrotic processes that lead to clinical disease. We report on vaccine design and the observed immunotherapeutic effects including induction of a balanced TH1/TH2/TH17 immune response that leads to reduced parasite burdens and tissue pathology. Further, we report vaccine-linked chemotherapy, a dose sparing strategy to further reduce parasite burdens and tissue pathology. EXPERT OPINION Our vaccine-linked chemotherapeutic approach is a multimodal treatment strategy, addressing both the parasite persistence and the underlying deleterious host inflammatory and fibrotic responses that lead to cardiac dysfunction. In targeting treatment towards patients with chronic indeterminate or early determinate Chagas disease, this vaccine-linked chemotherapeutic approach will be highly economical and will reduce the global disease burden and deaths due to CCC.
Collapse
Affiliation(s)
- Kathryn M Jones
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Cristina Poveda
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Leroy Versteeg
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.,Cell Biology and Immunology Group, Wageningen University & Research, De Elst 1, 6708 WD Wageningen, The Netherlands
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Biology, Baylor University, Waco, Texas, United States of America
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.,Department of Biology, Baylor University, Waco, Texas, United States of America.,James A. Baker III Institute for Public Policy, Rice University, Houston, Texas, United States of America.,Hagler Institute for Advanced Study at Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
9
|
Vieira RDS, Nascimento MS, Noronha IH, Vasconcelos JRC, Benvenuti LA, Barber GN, Câmara NOS, Kalil J, Cunha-Neto E, Almeida RR. STING Signaling Drives Production of Innate Cytokines, Generation of CD8 + T Cells and Enhanced Protection Against Trypanosoma cruzi Infection. Front Immunol 2022; 12:775346. [PMID: 35095849 PMCID: PMC8795786 DOI: 10.3389/fimmu.2021.775346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022] Open
Abstract
A variety of signaling pathways are involved in the induction of innate cytokines and CD8+ T cells, which are major players in protection against acute Trypanosoma cruzi infection. Previous data have demonstrated that a TBK-1/IRF3-dependent signaling pathway promotes IFN-β production in response to Trypanosoma cruzi, but the role for STING, a main interactor of these proteins, remained to be addressed. Here, we demonstrated that STING signaling is required for production of IFN-β, IL-6, and IL-12 in response to Trypanosoma cruzi infection and that STING absence negatively impacts activation of IRF-dependent pathways in response to the parasite. We reported no significant activation of IRF-dependent pathways and cytokine expression in RAW264.7 macrophages in response to heat-killed trypomastigotes. In addition, we showed that STING is essential for T. cruzi DNA-mediated induction of IFN-β, IL-6, and IL-12 gene expression in RAW264.7 macrophages. We demonstrated that STING-knockout mice have significantly higher parasitemia from days 5 to 8 of infection and higher heart parasitism at day 13 after infection. Although we observed similar heart inflammatory infiltrates at day 13 after infection, IFN-β, IL-12, CXCL9, IFN-γ, and perforin gene expression were lower in the absence of STING. We also showed an inverse correlation between parasite DNA and the expression of CXCL9, IFN-γ, and perforin genes in the hearts of infected animals at day 13 after infection. Finally, we reported that STING signaling is required for splenic IFN-β and IL-6 expression early after infection and that STING deficiency results in lower numbers of splenic parasite-specific IFN-γ and IFN-γ/perforin-producing CD8+ T cells, indicating a pivotal role for STING signaling in immunity to Trypanosoma cruzi.
Collapse
Affiliation(s)
- Raquel de Souza Vieira
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marilda Savoia Nascimento
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Isaú Henrique Noronha
- Laboratório de Vacinas Recombinantes, Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| | | | - Luiz Alberto Benvenuti
- Divisão de Patologia, Instituto do Coração (INCOR), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Glen N Barber
- Department of Cell Biology, University of Miami, Miami, FL, United States
| | - Niels Olsen Saraiva Câmara
- Laboratório de Imunologia Experimental e Clínica, Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Laboratório de Imunologia de Transplantes, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto de Investigação em Imunologia (III), Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto de Investigação em Imunologia (III), Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Teixeira PC, Ducret A, Langen H, Nogoceke E, Santos RHB, Silva Nunes JP, Benvenuti L, Levy D, Bydlowski SP, Bocchi EA, Kuramoto Takara A, Fiorelli AI, Stolf NA, Pomeranzeff P, Chevillard C, Kalil J, Cunha-Neto E. Impairment of Multiple Mitochondrial Energy Metabolism Pathways in the Heart of Chagas Disease Cardiomyopathy Patients. Front Immunol 2021; 12:755782. [PMID: 34867990 PMCID: PMC8633876 DOI: 10.3389/fimmu.2021.755782] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
Chagas disease cardiomyopathy (CCC) is an inflammatory dilated cardiomyopathy occurring in 30% of the 6 million infected with the protozoan Trypanosoma cruzi in Latin America. Survival is significantly lower in CCC than ischemic (IC) and idiopathic dilated cardiomyopathy (DCM). Previous studies disclosed a selective decrease in mitochondrial ATP synthase alpha expression and creatine kinase activity in CCC myocardium as compared to IDC and IC, as well as decreased in vivo myocardial ATP production. Aiming to identify additional constraints in energy metabolism specific to CCC, we performed a proteomic study in myocardial tissue samples from CCC, IC and DCM obtained at transplantation, in comparison with control myocardial tissue samples from organ donors. Left ventricle free wall myocardial samples were subject to two-dimensional electrophoresis with fluorescent labeling (2D-DIGE) and protein identification by mass spectrometry. We found altered expression of proteins related to mitochondrial energy metabolism, cardiac remodeling, and oxidative stress in the 3 patient groups. Pathways analysis of proteins differentially expressed in CCC disclosed mitochondrial dysfunction, fatty acid metabolism and transmembrane potential of mitochondria. CCC patients’ myocardium displayed reduced expression of 22 mitochondrial proteins belonging to energy metabolism pathways, as compared to 17 in DCM and 3 in IC. Significantly, 6 beta-oxidation enzymes were reduced in CCC, while only 2 of them were down-regulated in DCM and 1 in IC. We also observed that the cytokine IFN-gamma, previously described with increased levels in CCC, reduces mitochondrial membrane potential in cardiomyocytes. Results suggest a major reduction of mitochondrial energy metabolism and mitochondrial dysfunction in CCC myocardium which may be in part linked to IFN-gamma. This may partially explain the worse prognosis of CCC as compared to DCM or IC.
Collapse
Affiliation(s)
- Priscila Camillo Teixeira
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Axel Ducret
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Hanno Langen
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Everson Nogoceke
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | | | - João Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto Nacional de Ciência e Tecnologia, INCT, iii- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Luiz Benvenuti
- Anatomical Pathology Division, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Debora Levy
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Edimar Alcides Bocchi
- Heart Failure Team, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Andréia Kuramoto Takara
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Alfredo Inácio Fiorelli
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Noedir Antonio Stolf
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pablo Pomeranzeff
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Christophe Chevillard
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto Nacional de Ciência e Tecnologia, INCT, iii- Institute for Investigation in Immunology, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto Nacional de Ciência e Tecnologia, INCT, iii- Institute for Investigation in Immunology, São Paulo, Brazil
| |
Collapse
|
11
|
Nunes JPS, Andrieux P, Brochet P, Almeida RR, Kitano E, Honda AK, Iwai LK, Andrade-Silva D, Goudenège D, Alcântara Silva KD, Vieira RDS, Levy D, Bydlowski SP, Gallardo F, Torres M, Bocchi EA, Mano M, Santos RHB, Bacal F, Pomerantzeff P, Laurindo FRM, Teixeira PC, Nakaya HI, Kalil J, Procaccio V, Chevillard C, Cunha-Neto E. Co-Exposure of Cardiomyocytes to IFN-γ and TNF-α Induces Mitochondrial Dysfunction and Nitro-Oxidative Stress: Implications for the Pathogenesis of Chronic Chagas Disease Cardiomyopathy. Front Immunol 2021; 12:755862. [PMID: 34867992 PMCID: PMC8632642 DOI: 10.3389/fimmu.2021.755862] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
Infection by the protozoan Trypanosoma cruzi causes Chagas disease cardiomyopathy (CCC) and can lead to arrhythmia, heart failure and death. Chagas disease affects 8 million people worldwide, and chronic production of the cytokines IFN-γ and TNF-α by T cells together with mitochondrial dysfunction are important players for the poor prognosis of the disease. Mitochondria occupy 40% of the cardiomyocytes volume and produce 95% of cellular ATP that sustain the life-long cycles of heart contraction. As IFN-γ and TNF-α have been described to affect mitochondrial function, we hypothesized that IFN-γ and TNF-α are involved in the myocardial mitochondrial dysfunction observed in CCC patients. In this study, we quantified markers of mitochondrial dysfunction and nitro-oxidative stress in CCC heart tissue and in IFN-γ/TNF-α-stimulated AC-16 human cardiomyocytes. We found that CCC myocardium displayed increased levels of nitro-oxidative stress and reduced mitochondrial DNA as compared with myocardial tissue from patients with dilated cardiomyopathy (DCM). IFN-γ/TNF-α treatment of AC-16 cardiomyocytes induced increased nitro-oxidative stress and decreased the mitochondrial membrane potential (ΔΨm). We found that the STAT1/NF-κB/NOS2 axis is involved in the IFN-γ/TNF-α-induced decrease of ΔΨm in AC-16 cardiomyocytes. Furthermore, treatment with mitochondria-sparing agonists of AMPK, NRF2 and SIRT1 rescues ΔΨm in IFN-γ/TNF-α-stimulated cells. Proteomic and gene expression analyses revealed that IFN-γ/TNF-α-treated cells corroborate mitochondrial dysfunction, transmembrane potential of mitochondria, altered fatty acid metabolism and cardiac necrosis/cell death. Functional assays conducted on Seahorse respirometer showed that cytokine-stimulated cells display decreased glycolytic and mitochondrial ATP production, dependency of fatty acid oxidation as well as increased proton leak and non-mitochondrial oxygen consumption. Together, our results suggest that IFN-γ and TNF-α cause direct damage to cardiomyocytes’ mitochondria by promoting oxidative and nitrosative stress and impairing energy production pathways. We hypothesize that treatment with agonists of AMPK, NRF2 and SIRT1 might be an approach to ameliorate the progression of Chagas disease cardiomyopathy.
Collapse
Affiliation(s)
- João Paulo Silva Nunes
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil.,INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Pauline Andrieux
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Pauline Brochet
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Rafael Ribeiro Almeida
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Eduardo Kitano
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - André Kenji Honda
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Leo Kei Iwai
- Laboratório Especial de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - Débora Andrade-Silva
- Laboratório Especial de Toxinologia Aplicada, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Instituto Butantan, São Paulo, Brazil
| | - David Goudenège
- Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
| | - Karla Deysiree Alcântara Silva
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Raquel de Souza Vieira
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Débora Levy
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Paulo Bydlowski
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Frédéric Gallardo
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Magali Torres
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Edimar Alcides Bocchi
- Heart Failure Team, Heart Institute (Incor) Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Laboratory, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | | | - Fernando Bacal
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pablo Pomerantzeff
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Priscila Camillo Teixeira
- Translational Research Sciences, Pharma Research and Early Development F. Hoffmann-La Roche, Basel, Switzerland
| | | | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Vincent Procaccio
- MitoLab, UMR CNRS 6015-INSERM U1083, Université d'Angers, Angers, France
| | - Christophe Chevillard
- INSERM, UMR_1090, Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Institut MarMaRa, Marseille, France
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,iii-Institute for Investigation in Immunology, Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| |
Collapse
|
12
|
Pedra-Rezende Y, Barbosa JMC, Bombaça ACS, Dantas-Pereira L, Gibaldi D, Vilar-Pereira G, Dos Santos HAM, Ramos IP, Silva-Gomes NL, Moreira OC, Lannes-Vieira J, Menna-Barreto RFS. Physical Exercise Promotes a Reduction in Cardiac Fibrosis in the Chronic Indeterminate Form of Experimental Chagas Disease. Front Immunol 2021; 12:712034. [PMID: 34804007 PMCID: PMC8599157 DOI: 10.3389/fimmu.2021.712034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 10/15/2021] [Indexed: 01/14/2023] Open
Abstract
Chagas disease (CD), caused by the protozoan Trypanosoma cruzi, is a neglected tropical disease and a health problem in Latin America. Etiological treatment has limited effectiveness in chronic CD; thus, new therapeutic strategies are required. The practice of physical exercises has been widely advocated to improve the quality of life of CD patients. The most frequent clinical CD manifestation is the chronic indeterminate form (CIF), and the effect of physical exercises on disease progression remains unknown. Here, in a CIF model, we aimed to evaluate the effect of physical exercises on cardiac histological, parasitological, mitochondrial, and oxidative metabolism, electro and echocardiographic profiles, and immunological features. To establish a CIF model, BALB/c and C57BL/6 mice were infected with 100 and 500 trypomastigotes of the Y T. cruzi strain. At 120 days postinfection (dpi), all mouse groups showed normal PR and corrected QT intervals and QRS complexes. Compared to BALB/c mice, C57BL/6 mice showed a lower parasitemia peak, mortality rate, and less intense myocarditis. Thus, C57BL/6 mice infected with 500 parasites were used for subsequent analyses. At 120 dpi, a decrease in cardiac mitochondrial oxygen consumption and an increase in reactive oxygen species (ROS) were detected. When we increased the number of analyzed mice, a reduced heart rate and slightly prolonged corrected QT intervals were detected, at 120 and 150 dpi, which were then normalized at 180 dpi, thus characterizing the CIF. Y-infected mice were subjected to an exercise program on a treadmill for 4 weeks (from 150 to 180 dpi), five times per week in a 30–60-min daily training session. At 180 dpi, no alterations were detected in cardiac mitochondrial and oxidative metabolism, which were not affected by physical exercises, although ROS production increased. At 120 and 180 dpi, comparing infected and non-infected mice, no differences were observed in the levels of plasma cytokines, indicating that a crucial biomarker of the systemic inflammatory profile was absent and not affected by exercise. Compared with sedentary mice, trained Y-infected mice showed similar parasite loads and inflammatory cells but reduced cardiac fibrosis. Therefore, our data show that physical exercises promote beneficial changes that may prevent CD progression.
Collapse
Affiliation(s)
- Yasmin Pedra-Rezende
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Laboratório de Biologia das Interações, Instituto Oswaldo Cruz Oswaldo Cruz, Fundação, Rio de Janeiro, Brazil
| | - Juliana M C Barbosa
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ana Cristina S Bombaça
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Luiza Dantas-Pereira
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Laboratório de Biologia das Interações, Instituto Oswaldo Cruz Oswaldo Cruz, Fundação, Rio de Janeiro, Brazil
| | - Daniel Gibaldi
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz Oswaldo Cruz, Fundação, Rio de Janeiro, Brazil
| | - Glaucia Vilar-Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz Oswaldo Cruz, Fundação, Rio de Janeiro, Brazil.,Instituto Brasileiro de Medicina de Reabilitação, Rio de Janeiro, Brazil
| | - Hílton Antônio Mata Dos Santos
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratório de Análise e Desenvolvimento de Inibidores Enzimáticos e Laboratório Multiusuário de Análises por RMN, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isalira Peroba Ramos
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natália Lins Silva-Gomes
- Plataforma de PCR em Tempo Real RPT09A, Laboratório de Biologia Molecular de Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Otacilio C Moreira
- Plataforma de PCR em Tempo Real RPT09A, Laboratório de Biologia Molecular de Doenças Endêmicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz Oswaldo Cruz, Fundação, Rio de Janeiro, Brazil
| | - Rubem F S Menna-Barreto
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Silva NSDL, Orikaza CM, de Santana FR, Dos Santos LA, Salu BR, Oliva MLV, Sinigaglia RDC, Mortara RA. Interleukin-9 in Immunopathology of Trypanosoma cruzi Experimental Infection. Front Cell Infect Microbiol 2021; 11:756521. [PMID: 34722343 PMCID: PMC8554238 DOI: 10.3389/fcimb.2021.756521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/27/2021] [Indexed: 11/27/2022] Open
Abstract
Chagas’ disease is a parasitosis caused by Trypanosoma cruzi, which affects approximately 8 million people worldwide. The balance between pro- and anti-inflammatory cytokines produced during immunological responses contributes to disease prognosis and progression. Parasite tissue persistence can induce chronic inflammatory stimuli, which can cause long-term tissue injury and fibrosis. Chronic Chagas’ patients exhibit increased levels of interleukin (IL)-9, an important cytokine in the regulation of inflammatory and fibrogenic processes. Data on the role of IL-9 in other pathologies are sometimes contradictory, and few studies have explored this cytokine’s influence in Chagas’ disease pathology. Hence, the aim of this study was to evaluate the role of IL-9 in the progression of T. cruzi infection in vivo and in vitro. In vitro infection demonstrated that IL-9 reduced the number of infected cells and decreased the multiplication of intracellular amastigotes in both C2C12 myoblasts and bone marrow-derived macrophages. In myoblasts, the increased production of nitric oxide (NO) was essential for reduced parasite multiplication, whereas macrophage responses resulted in increased IL-6 and reduced TGF-β levels, indicating that parasite growth restriction mechanisms induced by IL-9 were cell-type specific. Experimental infection of BALB/c mice with T. cruzi trypomastigotes of the Y strain implicated a major role of IL-9 during the chronic phase, as increased Th9 and Tc9 cells were detected among splenocytes; higher levels of IL-9 in these cell populations and increased cardiac IL-9 levels were detected compared to those of uninfected mice. Moreover, rIL9 treatment decreased serum IL-12, IL-6, and IL-10 levels and cardiac TNF-α levels, possibly attempting to control the inflammatory response. IL-9 neutralization increased cardiac fibrosis, synthesis of collagens I and III, and mastocyte recruitment in BALB/c heart tissue during the chronic phase. In conclusion, our data showed that IL-9 reduced the invasion and multiplication of T. cruzi in vitro, in both myoblasts and macrophages, favoring disease control through cell-specific mechanisms. In vivo, IL-9 was elevated during experimental chronic infection in BALB/c mice, and this cytokine played a protective role in the immunopathological response during this phase by controlling cardiac fibrosis and proinflammatory cytokine production.
Collapse
Affiliation(s)
- Nadjania Saraiva de Lira Silva
- Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Cristina Mary Orikaza
- Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Fabiana Rodrigues de Santana
- Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Luana Aguiar Dos Santos
- Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Bruno Ramos Salu
- Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Maria Luiza Vilela Oliva
- Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Rita de Cássia Sinigaglia
- Electronic Microscopy Center, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| | - Renato Arruda Mortara
- Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Neves EGA, Koh CC, Padilha da Silva JL, Passos LSA, Villani FNA, Dos Santos JSC, Menezes CAS, Silva VR, Tormin JPAS, Evangelista GFB, Carvalho ATD, Rocha MODC, Nascimento B, Gollob KJ, Nunes MDCP, Dutra WO. Systemic cytokines, chemokines and growth factors reveal specific and shared immunological characteristics in infectious cardiomyopathies. Cytokine 2021; 148:155711. [PMID: 34592495 DOI: 10.1016/j.cyto.2021.155711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022]
Abstract
Heart disease is a major cause of death worldwide. Chronic Chagas cardiomyopathy (CCC) caused by infection with Trypanosoma cruzi leading to high mortality in adults, and rheumatic heart disease (RHD), resulting from infection by Streptococcus pyogenes affecting mainly children and young adults, are amongst the deadliest heart diseases in low-middle income countries. Despite distinct etiology, the pathology associated with both diseases is a consequence of inflammation. Here we compare systemic immune profile in patients with these cardiopathies, to identify particular and common characteristics in these infectious heart diseases. We evaluated the expression of 27 soluble factors, employing single and multivariate analysis combined with machine-learning approaches. We observed that, while RHD and CCC display higher levels of circulating mediators than healthy individuals, CCC is associated with stronger immune activation as compared to RHD. Despite distinct etiologies, univariate analysis showed that expression of TNF, IL-17, IFN-gamma, IL-4, CCL4, CCL3, CXCL8, CCL11, CCL2, PDGF-BB were similar between CCC and RHD, consistent with their inflammatory nature. Network analysis revealed common inflammatory pathways between CCC and RHD, while highlighting the broader reach of the inflammatory response in CCC. The final multivariate model showed a 100% discrimination power for the combination of the cytokines IL-12p70, IL-1Ra, IL-4, and IL-7 between CCC and RHD groups. Thus, while clear immunological distinctions were identified between CCC and RHD, similarities indicate shared inflammatory pathways in these infectious heart diseases. These results contribute to understanding the pathogenesis of CCC and RHD and may impact the design of immune-based therapies for these and other inflammatory cardiopathies that may also share immunological characteristics.
Collapse
Affiliation(s)
- Eula G A Neves
- Cell-cell Interactions Laboratory, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carolina C Koh
- Cell-cell Interactions Laboratory, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Lívia S A Passos
- Cell-cell Interactions Laboratory, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Cristiane A S Menezes
- Department of Clinical and Toxicological Analysis, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vicente R Silva
- Graduate Program in Infectology and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Julia P A S Tormin
- Graduate Program in Infectology and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Manoel Otávio da Costa Rocha
- Graduate Program in Infectology and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bruno Nascimento
- Graduate Program in Infectology and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Kenneth John Gollob
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, BA, Brazil
| | - Maria do Carmo P Nunes
- Graduate Program in Infectology and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walderez O Dutra
- Cell-cell Interactions Laboratory, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, BA, Brazil.
| |
Collapse
|
15
|
Queiroga TBD, Pereira NDS, da Silva DD, Andrade CDM, de Araújo Júnior RF, Brito CRDN, Galvão LMDC, da Câmara ACJ, Nascimento MSL, Guedes PMM. Virulence of Trypanosoma cruzi Strains Is Related to the Differential Expression of Innate Immune Receptors in the Heart. Front Cell Infect Microbiol 2021; 11:696719. [PMID: 34336720 PMCID: PMC8321543 DOI: 10.3389/fcimb.2021.696719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/25/2021] [Indexed: 11/13/2022] Open
Abstract
Resistance or susceptibility to T. cruzi infection is dependent on the host immunological profile. Innate immune receptors, such as Toll-like receptors (TLRs/TLR2, TLR4, TLR7, and TLR9) and Nod-like receptors (NLRs/NOD1 and NLRP3 inflammasome) are involved with the resistance against acute experimental T. cruzi infection. Here, we evaluated the impact of T. cruzi virulence on the expression of innate immune receptors and its products in mice. For that, we used six T. cruzi strains/isolates that showed low (AM64/TcIV and 3253/Tc-V), medium (PL1.10.14/TcIII and CL/TcVI), or high (Colombian/Tc-I and Y/TcII) virulence and pathogenicity to the vertebrate host and belonging to the six discrete typing units (DTUs)—TcI to TcVI. Parasitemia, mortality, and myocarditis were evaluated and correlated to the expression of TLRs, NLRs, adapter molecules, cytokines, and iNOS in myocardium by real time PCR. Cytokines (IL-1β, IL-12, TNF-α, and IFN-γ) were quantified in sera 15 days after infection. Our data indicate that high virulent strains of T. cruzi, which generate high parasitemia, severe myocarditis, and 100% mortality in infected mice, inhibit the expression of TLR2, TLR4, TLR9, TRIF, and Myd88 transcripts, leading to a low IL-12 production, when compared to medium and low virulent T. cruzi strains. On the other hand, the high virulent T. cruzi strains induce the upregulation of NLRP3, caspase-1, IL-1β, TNF-α, and iNOS mRNA in heart muscle, compared to low and medium virulent strains, which may contribute to myocarditis and death. Moreover, high virulent strains induce higher levels of IL-1β and TNF-α in sera compared to less virulent parasites. Altogether the data indicate that differential TLR and NLR expression in heart muscle is correlated with virulence and pathogenicity of T cruzi strains. A better knowledge of the immunological mechanisms involved in resistance to T. cruzi infection is important to understand the natural history of Chagas disease, can lead to identification of immunological markers and/or to serve as a basis for alternative therapies.
Collapse
Affiliation(s)
| | - Nathalie de Sena Pereira
- Graduate Program Health and Biological Sciences, Federal University of Vale do São Francisco, Petrolina, Brazil
| | - Denis Dantas da Silva
- Graduate Program Parasitary Biology, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Raimundo Fernandes de Araújo Júnior
- Laboratory of Investigation of the Inflammation and Cancer (LAICI)/Department of Morphology, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | | | | | | | - Paulo Marcos Matta Guedes
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
16
|
Rare Pathogenic Variants in Mitochondrial and Inflammation-Associated Genes May Lead to Inflammatory Cardiomyopathy in Chagas Disease. J Clin Immunol 2021; 41:1048-1063. [PMID: 33660144 PMCID: PMC8249271 DOI: 10.1007/s10875-021-01000-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/15/2021] [Indexed: 01/21/2023]
Abstract
Abstract Cardiomyopathies are an important cause of heart failure and sudden cardiac death. Little is known about the role of rare genetic variants in inflammatory cardiomyopathy. Chronic Chagas disease cardiomyopathy (CCC) is an inflammatory cardiomyopathy prevalent in Latin America, developing in 30% of the 6 million patients chronically infected by the protozoan Trypanosoma cruzi, while 60% remain free of heart disease (asymptomatic (ASY)). The cytokine interferon-γ and mitochondrial dysfunction are known to play a major pathogenetic role. Chagas disease provides a unique model to probe for genetic variants involved in inflammatory cardiomyopathy. Methods We used whole exome sequencing to study nuclear families containing multiple cases of Chagas disease. We searched for rare pathogenic variants shared by all family members with CCC but absent in infected ASY siblings and in unrelated ASY. Results We identified heterozygous, pathogenic variants linked to CCC in all tested families on 22 distinct genes, from which 20 were mitochondrial or inflammation-related – most of the latter involved in proinflammatory cytokine production. Significantly, incubation with IFN-γ on a human cardiomyocyte line treated with an inhibitor of dihydroorotate dehydrogenase brequinar (enzyme showing a loss-of-function variant in one family) markedly reduced mitochondrial membrane potential (ΔψM), indicating mitochondrial dysfunction. Conclusion Mitochondrial dysfunction and inflammation may be genetically determined in CCC, driven by rare genetic variants. We hypothesize that CCC-linked genetic variants increase mitochondrial susceptibility to IFN-γ-induced damage in the myocardium, leading to the cardiomyopathy phenotype in Chagas disease. This mechanism may also be operative in other inflammatory cardiomyopathies. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-021-01000-y.
Collapse
|
17
|
Laugier L, Ferreira LRP, Ferreira FM, Cabantous S, Frade AF, Nunes JP, Ribeiro RA, Brochet P, Teixeira PC, Santos RHB, Bocchi EA, Bacal F, Cândido DDS, Maso VE, Nakaya HI, Kalil J, Cunha-Neto E, Chevillard C. miRNAs may play a major role in the control of gene expression in key pathobiological processes in Chagas disease cardiomyopathy. PLoS Negl Trop Dis 2020; 14:e0008889. [PMID: 33351798 PMCID: PMC7787679 DOI: 10.1371/journal.pntd.0008889] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 01/06/2021] [Accepted: 10/14/2020] [Indexed: 01/19/2023] Open
Abstract
Chronic Chagas disease cardiomyopathy (CCC), an especially aggressive inflammatory dilated cardiomyopathy caused by lifelong infection with the protozoan Trypanosoma cruzi, is a major cause of cardiomyopathy in Latin America. Although chronic myocarditis may play a major pathogenetic role, little is known about the molecular mechanisms responsible for its severity. The aim of this study is to study the genes and microRNAs expression in tissues and their connections in regards to the pathobiological processes. To do so, we integrated for the first time global microRNA and mRNA expression profiling from myocardial tissue of CCC patients employing pathways and network analyses. We observed an enrichment in biological processes and pathways associated with the immune response and metabolism. IFNγ, TNF and NFkB were the top upstream regulators. The intersections between differentially expressed microRNAs and differentially expressed target mRNAs showed an enrichment in biological processes such as Inflammation, inflammation, Th1/IFN-γ-inducible genes, fibrosis, hypertrophy, and mitochondrial/oxidative stress/antioxidant response. MicroRNAs also played a role in the regulation of gene expression involved in the key cardiomyopathy-related processes fibrosis, hypertrophy, myocarditis and arrhythmia. Significantly, a discrete number of differentially expressed microRNAs targeted a high number of differentially expressed mRNAs (>20) in multiple processes. Our results suggest that miRNAs orchestrate expression of multiple genes in the major pathophysiological processes in CCC heart tissue. This may have a bearing on pathogenesis, biomarkers and therapy. Chronic Chagas disease cardiomyopathy (CCC), an aggressive dilated cardiomyopathy caused by Trypanosoma cruzi, is a major cause of cardiomyopathy in Latin America. Little is known about the molecular mechanisms responsible for its severity. Authors study the possible role of microRNAs in the regulation of gene expression in relevant pathways and pathobiological processes. Differentially expressed genes (DEGs) and differentially expressed miRNAs (DEMs) -small RNAs that can regulate gene expression—associated to severe cardiomyopathy development. The inflammatory mediator Interferon-γ was the most likely inducer of gene expression in CCC, and most genes belonged to the immune response, fibrosis, hypertrophy and mitochondrial metabolism. A discrete number of differentially expressed mRNAs targeted a high number of differentially expressed mRNAs in multiple processes. Moreover, several pathways had multiple targets regulated by microRNAs, suggesting synergic effect. Results suggest that microRNAs orchestrate expression of multiple genes in the major pathophysiological processes in CCC heart tissue.
Collapse
Affiliation(s)
- Laurie Laugier
- Aix Marseille Université, Génétique et Immunologie des Maladies Parasitaires, Unité Mixte de Recherche S906, Marseille, France; INSERM, U906, Marseille, France
| | - Ludmila Rodrigues Pinto Ferreira
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Frederico Moraes Ferreira
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Sandrine Cabantous
- Aix Marseille Université, Génétique et Immunologie des Maladies Parasitaires, Unité Mixte de Recherche S906, Marseille, France; INSERM, U906, Marseille, France
| | - Amanda Farage Frade
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Joao Paulo Nunes
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Rafael Almeida Ribeiro
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Pauline Brochet
- Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Inserm, INSERM, UMR_1090, Marseille, France
| | - Priscila Camillo Teixeira
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | | | - Edimar A Bocchi
- Division of Transplantation, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Fernando Bacal
- Division of Transplantation, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Darlan da Silva Cândido
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Vanessa Escolano Maso
- Department of Pathophysiology and Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Helder I Nakaya
- Department of Pathophysiology and Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.,Scientific Platform Pasteur, University of São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor), University of São Paulo, School of Medicine, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo, School of Medicine, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Christophe Chevillard
- Aix Marseille Université, TAGC Theories and Approaches of Genomic Complexity, Inserm, INSERM, UMR_1090, Marseille, France
| |
Collapse
|
18
|
CD8low T cells expanded following acute Trypanosoma cruzi infection and benznidazole treatment are a relevant subset of IFN-γ producers. PLoS Negl Trop Dis 2020; 14:e0008969. [PMID: 33347472 PMCID: PMC7785226 DOI: 10.1371/journal.pntd.0008969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/05/2021] [Accepted: 11/10/2020] [Indexed: 11/30/2022] Open
Abstract
CD8 T cells are regarded as pivotal players in both immunoprotection and immunopathology following Trypanosoma cruzi infection. Previously, we demonstrated the expansion of CD8+ T lymphocytes in the spleen of T. cruzi-infected mice under treatment with benznidazole (N-benzyl-2-nitroimidazole acetamide; Bz), a drug available for clinical therapy. This finding underlies the concept that the beneficial effects of Bz on controlling acute T. cruzi infection are related to a synergistic process between intrinsic trypanocidal effect and indirect triggering of the active immune response. In the present study, we particularly investigated the effect of Bz treatment on the CD8+ T cell subset following T. cruzi infection. Herein we demonstrated that, during acute T. cruzi infection, Bz treatment reduces and abbreviates the parasitemia, but maintains elevated expansion of CD8+ T cells. Within this subset, a remarkable group of CD8low cells was found in both Bz-treated and non-treated infected mice. In Bz-treated mice, early pathogen control paralleled the lower frequency of recently activated CD8low cells, as ascertained by CD69 expression. However, the CD8low subset sustains significant levels of CD44highCD62Llow and CD62LlowT-bethigh effector memory T cells, in both Bz-treated and non-treated infected mice. These CD8low cells also comprise the main group of spontaneous interferon (IFN)-γ-producing CD8+ T cells. Interestingly, following in vitro anti-CD3/CD28 stimulation, CD8+ T cells from Bz-treated T. cruzi-infected mice exhibited higher frequency of IFN-γ+ cells, which bear mostly a CD8low phenotype. Altogether, our results point to the marked presence of CD8low T cells that arise during acute T. cruzi infection, with Bz treatment promoting their significant expansion along with a potential effector program for high IFN-γ production. Chagas disease is a neglected illness caused by the protozoan Trypanosoma cruzi, which affects 6 to 7 million people worldwide. The current treatment for acutely-infected patients is mostly limited to the benznidazole (Bz) drug, reaching up to 80% of cure. It has been proposed that Bz therapy efficacy involves both trypanocidal and immunonodulatory effects. In this sense, we previously suggested that CD8+ T cells, highly expanded after Bz treatment of acute T. cruzi-infected mice, might play a particular role in parasite control. Here, by further investigating those expanded CD8+ T cells, we observed that they bear a clear-cut effector phenotype and that a significant part of them stand out as a subpopulation bearing low levels of CD8 on their surface. Interestingly, besides the evident parasite control, Bz-treated mice sustain a group of effector CD8low cells with spontaneous IFN-γ production. Moreover, in vitro-stimulated CD8+ T cells, sorted from infected and Bz-treated mice, present a relevant group of IFN-γ+ cells with a CD8low phenotype. Altogether, our data indicate the particular subset of CD8low cells as potentially contributing for a sustained protective immunity in T. cruzi-infected animals under Bz therapy.
Collapse
|
19
|
Gomes Dos Santos A, Watanabe EH, Ferreira DT, Oliveira J, Nakanishi ÉS, Oliveira CS, Bocchi E, Novaes CTG, Cruz F, Carvalho NB, Sato PK, Yamashiro-Kanashiro EH, Pontillo A, de Freitas VLT, Onuchic LF, Shikanai-Yasuda MA. A Specific IL6 Polymorphic Genotype Modulates the Risk of Trypanosoma cruzi Parasitemia While IL18, IL17A, and IL1B Variant Profiles and HIV Infection Protect Against Cardiomyopathy in Chagas Disease. Front Immunol 2020; 11:521409. [PMID: 33193300 PMCID: PMC7642879 DOI: 10.3389/fimmu.2020.521409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background Chagas disease caused by Trypanosoma cruzi (T. cruzi) affects approximately six million individuals worldwide. Clinical manifestations are expected to occur due to the parasite persistence and host immune response. Herein we investigated potential associations between IL1B, IL6, IL17A, or IL18 polymorphism profiles and cardiomyopathy or T. cruzi parasitemia, as well as the impact of HIV infection on cardiopathy. Methods Two hundred twenty-six patients and 90 control individuals were analyzed. IL1B rs1143627 T>C, IL6 rs1800795 C>G, IL17A rs2275913 G>A, IL18 rs187238 C>G, and IL18 rs1946518 C>A SNVs were analyzed by real-time PCR and T. cruzi parasitemia by PCR. Results Our data revealed association between a cytokine gene polymorphism and parasitemia never previously reported. The IL6 rs1800795 CG genotype lowered the risk of positive parasitemia (OR = 0.45, 95% CI 0.24–0.86, P = 0.015). Original findings included associations between IL17A rs2275913 AA and IL18 s1946518 AA genotypes with decreased risk of developing cardiomyopathy (OR = 0.27, 95% CI 0.07–0.97, P = 0.044; and OR = 0.35, 95% CI 0.14–0.87, P = 0.023, respectively). IL18 rs1946518 AA and IL1B rs1143627 TC were associated with reduced risk for cardiomyopathy severity, including NYHA (New York Heart Association) class ≥ 2 (OR = 0.21, 95% CI 0.06–0.68, P = 0.009; and OR = 0.48, 95% CI 0.24–0.95, P = 0.036, respectively) and LVEF (left ventricular ejection fraction) <45% for IL18 rs1946518 AA (OR = 0.22, 95% CI 0.05–0.89, P = 0.034). A novel, unexpected protective effect of HIV infection against development/progression of cardiomyopathy was identified, based on a lower risk of developing cardiopathy (OR = 0.48, 95% CI 0.23–0.96, P = 0.039), NYHA class ≥ 2 (OR = 0.15, 95% CI 0.06–0.39, P < 0.001), and LVEF < 45% (OR = 0.03, 95% CI 0.00–0.25, P = 0.001). Digestive involvement was negatively associated with NYHA ≥ 2 and LVEF < 45% (OR = 0.20, 95% CI 0.09–0.47, P < 0.001; and OR = 0.24, 95% CI 0.09–0.62, P = 0.004, respectively). Conclusions Our data support a protective role of IL17A AA, IL18 AA, and IL1B TC genotypes against development/progression of cardiomyopathy and a modulatory effect of the IL6 CG genotype on the risk of parasitemia in Chagas disease. Notably, HIV infection was shown to protect against development/progression of cardiopathy, potentially associated with a synergistic effect of HIV and highly active antiretroviral therapy (HAART), attenuating a Th1-mediated response in the myocardium. This proposed hypothesis requires confirmation, however, in larger and more comprehensive future studies.
Collapse
Affiliation(s)
- Alexandra Gomes Dos Santos
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Elieser Hitoshi Watanabe
- Department of Medicine, Divisions of Molecular Medicine and Nephrology, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Daiane Tomomi Ferreira
- Laboratory of Immunology (LIM 48), Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Jamille Oliveira
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Érika Shimoda Nakanishi
- Laboratory of Immunology (LIM 48), Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Claudia Silva Oliveira
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Edimar Bocchi
- Heart Institute, Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | | | - Fatima Cruz
- Heart Institute, Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Noemia Barbosa Carvalho
- Division of Infectious Diseases, Hospital das Clinicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Paula Keiko Sato
- Laboratory of Immunology (LIM 48), Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Edite Hatsumi Yamashiro-Kanashiro
- Laboratory of Immunology (LIM 48), Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil.,Instituto de Medicina Tropical, University of São Paulo, São Paulo, Brazil
| | - Alessandra Pontillo
- Departament of Immunology, Instituto de Ciências Biomédicas (ICB), University of São Paulo, São Paulo, Brazil
| | - Vera Lucia Teixeira de Freitas
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil.,Laboratory of Immunology (LIM 48), Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Luiz Fernando Onuchic
- Department of Medicine, Divisions of Molecular Medicine and Nephrology, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | - Maria Aparecida Shikanai-Yasuda
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil.,Laboratory of Immunology (LIM 48), Hospital das Clínicas, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Frade-Barros AF, Ianni BM, Cabantous S, Pissetti CW, Saba B, Lin-Wang HT, Buck P, Marin-Neto JA, Schmidt A, Dias F, Hirata MH, Sampaio M, Fragata A, Pereira AC, Donadi E, Rodrigues V, Kalil J, Chevillard C, Cunha-Neto E. Polymorphisms in Genes Affecting Interferon-γ Production and Th1 T Cell Differentiation Are Associated With Progression to Chagas Disease Cardiomyopathy. Front Immunol 2020; 11:1386. [PMID: 32733459 PMCID: PMC7358543 DOI: 10.3389/fimmu.2020.01386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Chagas disease, caused by the protozoan Trypanosoma cruzi, is endemic in Latin America. Thirty percent of infected individuals develop chronic Chagas cardiomyopathy (CCC), an inflammatory dilated cardiomyopathy that is the most important clinical consequence of T. cruzi infection, while the others remain asymptomatic (ASY). IFN-γ and IFN-γ-producing Th1-type T cells are increased in peripheral blood and CCC myocardium as compared to ASY patients, while the Th1-antagonizing cytokine IL-10 is more expressed in ASY patients. Importantly IFN-γ-producing Th1-type T cells are the most frequent cytokine-producing T cell subset in CCC myocardium, while expression of Th1-antagonizing cytokines IL-10 and IL-4 is unaltered. The control of IFN-γ production by Th1-type T cells may be a key event for progression toward CCC. A genetic component to disease progression was suggested by the familial aggregation of cases and the association of gene polymorphisms with CCC development. We here investigate the role of gene polymorphisms (SNPs) in several genes involved in the control of IFN-γ production and Th1 T cell differentiation in CCC development. Methods: We studied a Brazilian population including 315 CCC cases and 118 ASY subjects. We assessed 35 Tag SNPs designed to represent all the genetic information contained in the IL12B, IL10, IFNG, and IL4 genes. Results: We found 2 IL12 SNPs (rs2546893, rs919766) and a trend of association for a IL10 SNP (rs3024496) to be significantly associated with the ASY group. these associations were confirmed by multivariate analysis and allele tests. The rs919766C, 12rs2546893G, and rs3024496C alleles were associated to an increase risk to CCC development. Conclusions: Our data show that novel polymorphisms affecting IL12B and IL10, but not IFNG or IL4 genes play a role in genetic susceptibility to CCC development. This might indicate that the increased Th1 differentiation and IFN-γ production associated with CCC is genetically controlled.
Collapse
Affiliation(s)
- Amanda Farage Frade-Barros
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Aix-Marseille Université, INSERM, GIMP UMR_S906, Marseille, France.,Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, Brazil.,Bioengineering Program, Instituto Tecnológico, Universidade Brasil, São Paulo, Brazil
| | - Barbara Maria Ianni
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | | | - Cristina Wide Pissetti
- Laboratory of Immunology, Universidade Federal Do Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Bruno Saba
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Hui Tzu Lin-Wang
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Paula Buck
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - José Antonio Marin-Neto
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - André Schmidt
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Fabrício Dias
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Marcelo Sampaio
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Abílio Fragata
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Alexandre Costa Pereira
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - Eduardo Donadi
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Virmondes Rodrigues
- Laboratory of Immunology, Universidade Federal Do Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Jorge Kalil
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Bioengineering Program, Instituto Tecnológico, Universidade Brasil, São Paulo, Brazil
| | - Christophe Chevillard
- Aix Marseille Université, INSERM, TAGC Theories and Approaches of Genomic Complexity, UMR_1090, Marseille, France
| | - Edecio Cunha-Neto
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
21
|
de Araújo FF, Lima Torres KC, Viana Peixoto S, Pinho Ribeiro AL, Vaz Melo Mambrini J, Bortolo Rezende V, Lima Silva ML, Loyola Filho AI, Teixeira-Carvalho A, Lima-Costa MF, Martins-Filho OA. CXCL9 and CXCL10 display an age-dependent profile in Chagas patients: a cohort study of aging in Bambui, Brazil. Infect Dis Poverty 2020; 9:51. [PMID: 32393333 PMCID: PMC7216412 DOI: 10.1186/s40249-020-00663-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/17/2020] [Indexed: 12/18/2022] Open
Abstract
Background Chagas disease is endemic in Latin America and still represents an important public health problem in the region. Chronic cardiomyopathy is the most significant chronic form due to its association with morbidity and mortality. The last decade has seen increasing evidence that inflammatory cytokines and chemokines are responsible for the generation of inflammatory infiltrate and tissue damage, with chronic chagasic cardiomyopathy patients presenting a pro-inflammatory immune response. Although studies have evaluated the role of chemokines in experimental T. cruzi infection, few have addressed their systemic profile, especially for human infection and in aging populations. The present work aimed to use the data from a large population based study of older adults, conducted in an endemic area for Chagas disease, to examine the association between serum levels of cytokines and chemokines, T. cruzi infection and electrocardiogram (ECG) abnormality. Methods The present work evaluated serum levels of CCL2, CXCL9, CXCL10, CCL5, CXCL8, IL-1β, IL-6, TNF, IL-12 and IL-10 by Flow Cytometric Bead Array assay (CBA) and the results expressed in pg/ml. The baseline survey started in January 1st 1997, with 1284 participants of an aged population-based cohort. Participants signed an informed consent at baseline and at each subsequent visit and authorized death certificate and medical records verification. Results Our results demonstrated that Chagas disease patients had higher serum levels of CXCL9, CXCL10 and IL-1β and lower serum levels of CCL5 than non-infected subjects. Moreover, our data demonstrated that CXCL9 and CXCL10 increased in an age-dependent profile in Chagas disease patients. Conclusion Together, this study provided evidences that serum biomarkers increase along the age continuum and may have potential implications for establishing clinical management protocols and therapeutic intervention in Chagas disease patients.
Collapse
Affiliation(s)
- Fernanda Fortes de Araújo
- Integrated Research Group in Biomarkers, Rene Rachou Institute, Oswaldo Cruz Foundation, Avenida Augusto de Lima, 1715 - Barro Preto -, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Karen Cecília Lima Torres
- Integrated Research Group in Biomarkers, Rene Rachou Institute, Oswaldo Cruz Foundation, Avenida Augusto de Lima, 1715 - Barro Preto -, Belo Horizonte, Minas Gerais, 30190-002, Brazil.,José do Rosário Vellano University, UNIFENAS/BH, Belo Horizonte, Brazil
| | - Sérgio Viana Peixoto
- Center for Studies in Public Health and Aging, Rene Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | | | - Juliana Vaz Melo Mambrini
- Center for Studies in Public Health and Aging, Rene Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | - Vitor Bortolo Rezende
- Integrated Research Group in Biomarkers, Rene Rachou Institute, Oswaldo Cruz Foundation, Avenida Augusto de Lima, 1715 - Barro Preto -, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Maria Luiza Lima Silva
- Center for Studies in Public Health and Aging, Rene Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | - Antônio Ignácio Loyola Filho
- Center for Studies in Public Health and Aging, Rene Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | - Andréa Teixeira-Carvalho
- Integrated Research Group in Biomarkers, Rene Rachou Institute, Oswaldo Cruz Foundation, Avenida Augusto de Lima, 1715 - Barro Preto -, Belo Horizonte, Minas Gerais, 30190-002, Brazil
| | - Maria Fernanda Lima-Costa
- Center for Studies in Public Health and Aging, Rene Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Integrated Research Group in Biomarkers, Rene Rachou Institute, Oswaldo Cruz Foundation, Avenida Augusto de Lima, 1715 - Barro Preto -, Belo Horizonte, Minas Gerais, 30190-002, Brazil.
| |
Collapse
|
22
|
Cerbán FM, Stempin CC, Volpini X, Carrera Silva EA, Gea S, Motran CC. Signaling pathways that regulate Trypanosoma cruzi infection and immune response. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165707. [DOI: 10.1016/j.bbadis.2020.165707] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 02/07/2023]
|
23
|
de Paula Silva F, da Costa CMB, Pereira LM, Lessa DFS, Pitol DL, Issa JPM, do Prado Júnior JC, Abrahão AAC. Effects of ghrelin supplementation on the acute phase of Chagas disease in rats. Parasit Vectors 2019; 12:532. [PMID: 31706334 PMCID: PMC6842500 DOI: 10.1186/s13071-019-3787-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background Trypanosoma cruzi is the causative agent of Chagas disease, which is endemic to subtropical and tropical Americas. The disease treatment remains partially ineffective, involving therapies directed to the parasite as well as palliative strategies for the clinical manifestations. Therefore, novel candidates for disease control are necessary. Additionally, strategies based on parasite inhibition via specific targets and application of compounds which improve the immune response against the disease is welcomed. Ghrelin is a peptide hormone pointed as a substance with important cardioprotective, vasodilatory, anti-apoptotic, anti-oxidative and immune modulatory functions. The aims of this study were to evaluate the immunomodulatory effects of ghrelin in male Wistar rats infected with the Y strain of T. cruzi. Methods In order to delineate an immune response against T. cruzi mediated by ghrelin, we evaluated the following parameters: quantification of blood and cardiac parasites; analysis of cell markers (CD3+, CD8+, NK, NKT, CD45RA+, macrophage and RT1B+); nitric oxide (NO) production; lymphoproliferation assays; splenocyte apoptosis; and INF-γ, IL-12 and IL-6 quantification in sera. Results The animals infected with T. cruzi and supplemented with ghrelin demonstrated an upregulated pattern in macrophage and NO production, whereas an anti-inflammatory response was observed in T cells and cytokines. The low response against T. cruzi mediated by T cells probably contributed to a higher colonization of the cardiac tissue, when compared to infected groups. On the other side, the peptide decreased the inflammatory infiltration in cardiac tissue infected with T. cruzi. Conclusions Ghrelin demonstrated a dual function in animals infected with T. cruzi. Further studies, especially related to the decrease of cardiac tissue inflammation, are needed in order to determine the advantages of ghrelin supplementation in Chagas disease, mostly for populations from endemic areas.
Collapse
Affiliation(s)
- Ferdinando de Paula Silva
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-903, Brazil
| | - Cássia Mariana Bronzon da Costa
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-903, Brazil
| | - Luiz Miguel Pereira
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-903, Brazil
| | - Diego Fernando Silva Lessa
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-903, Brazil
| | - Dimitrius Leonardo Pitol
- Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto USP, University of São Paulo, Ribeirão Preto, 14040-904, Brazil
| | - João Paulo Mardegan Issa
- Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto USP, University of São Paulo, Ribeirão Preto, 14040-904, Brazil
| | - José Clóvis do Prado Júnior
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-903, Brazil
| | - Ana Amélia Carraro Abrahão
- Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, 14040-903, Brazil.
| |
Collapse
|
24
|
Silva TA, Ferreira LFDC, Pereira MCDS, Calvet CM. Differential Role of TGF-β in Extracellular Matrix Regulation During Trypanosoma cruzi-Host Cell Interaction. Int J Mol Sci 2019; 20:E4836. [PMID: 31569452 PMCID: PMC6801917 DOI: 10.3390/ijms20194836] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a determinant for inflammation and fibrosis in cardiac and skeletal muscle in Chagas disease. To determine its regulatory mechanisms, we investigated the response of Trypanosoma cruzi-infected cardiomyocytes (CM), cardiac fibroblasts (CF), and L6E9 skeletal myoblasts to TGF-β. Cultures of CM, CF, and L6E9 were infected with T. cruzi (Y strain) and treated with TGF-β (1-10 ng/mL, 1 h or 48 h). Fibronectin (FN) distribution was analyzed by immunofluorescence and Western blot (WB). Phosphorylated SMAD2 (PS2), phospho-p38 (p-p38), and phospho-c-Jun (p-c-Jun) signaling were evaluated by WB. CF and L6E9 showed an increase in FN from 1 ng/mL of TGF-β, while CM displayed FN modulation only after 10 ng/mL treatment. CF and L6E9 showed higher PS2 levels than CM, while p38 was less stimulated in CF than CM and L6E9. T. cruzi infection resulted in localized FN disorganization in CF and L6E9. T. cruzi induced an increase in FN in CF cultures, mainly in uninfected cells. Infected CF cultures treated with TGF-β showed a reduction in PS2 and an increase in p-p38 and p-c-Jun levels. Our data suggest that p38 and c-Jun pathways may be participating in the fibrosis regulatory process mediated by TGF-β after T. cruzi infection.
Collapse
Affiliation(s)
- Tatiana Araújo Silva
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-360, Brazil.
| | | | | | - Claudia Magalhães Calvet
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-360, Brazil.
| |
Collapse
|
25
|
Ambrosio LF, Insfran C, Volpini X, Acosta Rodriguez E, Serra HM, Quintana FJ, Cervi L, Motrán CC. Role of Aryl Hydrocarbon Receptor (AhR) in the Regulation of Immunity and Immunopathology During Trypanosoma cruzi Infection. Front Immunol 2019; 10:631. [PMID: 30984194 PMCID: PMC6450169 DOI: 10.3389/fimmu.2019.00631] [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: 12/21/2018] [Accepted: 03/08/2019] [Indexed: 12/15/2022] Open
Abstract
Resistance to Trypanosoma cruzi infection is dependent on a rapid induction of Th1-type and CD8+ T cell responses that should be promptly balanced to prevent immunopathology. T. cruzi-infected B6 mice are able to control parasite replication but show a limited expansion of Foxp3+regulatory T (Treg) cells that results in the accumulation of effector immune cells and the development of acute liver pathology. AhR is a ligand-activated transcription factor that promotes Treg cell development and suppression of pro-inflammatory cytokine production in dendritic cells, altering the course of adaptive immune response and the development of immunopathology. Here, we used different AhR-dependent activation strategies aiming to improve the Treg response, and B6 congenic mice carrying a mutant AhR variant with low affinity for its ligands (AhRd) to evaluate the role of AhR activation by natural ligands during experimental T. cruzi infection. The outcome of TCDD or 3-HK plus ITE treatments indicated that strong or weak AhR activation before or during T. cruzi infection was effective to regulate inflammation improving the Treg cell response and regularizing the ratio between CD4+ CD25- to Treg cells. However, AhR activation shifted the host-parasite balance to the parasite replication. Weak AhR activation resulted in Treg promotion while strong activation differentially modulated the susceptibility and resistance of cell death in activated T and Treg cells and the increase in TGF-β-producing Treg cells. Of note, T. cruzi-infected AhRd mice showed low levels of Treg cells associated with strong Th1-type response, low parasite burden and absence of liver pathology. These mice developed a Treg- and Tr1-independent mechanism of Th1 constriction showing increased levels of systemic IL-10 and IL-10-secreting CD4+ splenocytes. In addition, AhR activation induced by exogenous ligands had negative effects on the development of memory CD8+ T cell subsets while the lack/very weak activation in AhRd mice showed opposite results, suggesting that AhR ligation restricts the differentiation of memory CD8+T cell subsets. We propose a model in which a threshold of AhR activation exists and may explain how activation or inhibition of AhR-derived signals by infection/inflammation-induced ligands, therapeutic interventions or exposure to pollutants can modulate infections/diseases outcomes or vaccination efficacy.
Collapse
Affiliation(s)
- Laura Fernanda Ambrosio
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Constanza Insfran
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Ximena Volpini
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Eva Acosta Rodriguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Horacio Marcelo Serra
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,The Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| |
Collapse
|
26
|
Chevillard C, Nunes JPS, Frade AF, Almeida RR, Pandey RP, Nascimento MS, Kalil J, Cunha-Neto E. Disease Tolerance and Pathogen Resistance Genes May Underlie Trypanosoma cruzi Persistence and Differential Progression to Chagas Disease Cardiomyopathy. Front Immunol 2018; 9:2791. [PMID: 30559742 PMCID: PMC6286977 DOI: 10.3389/fimmu.2018.02791] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 11/13/2018] [Indexed: 01/01/2023] Open
Abstract
Chagas disease is caused by infection with the protozoan Trypanosoma cruzi and affects over 8 million people worldwide. In spite of a powerful innate and adaptive immune response in acute infection, the parasite evades eradication, leading to a chronic persistent infection with low parasitism. Chronically infected subjects display differential patterns of disease progression. While 30% develop chronic Chagas disease cardiomyopathy (CCC)—a severe inflammatory dilated cardiomyopathy—decades after infection, 60% of the patients remain disease-free, in the asymptomatic/indeterminate (ASY) form, and 10% develop gastrointestinal disease. Infection of genetically deficient mice provided a map of genes relevant for resistance to T. cruzi infection, leading to the identification of multiple genes linked to survival to infection. These include pathogen resistance genes (PRG) needed for intracellular parasite destruction, and genes involved in disease tolerance (protection against tissue damage and acute phase death—DTG). All identified DTGs were found to directly or indirectly inhibit IFN-γ production or Th1 differentiation. We hypothesize that the absolute need for DTG to control potentially lethal IFN-γ PRG activity leads to T. cruzi persistence and establishment of chronic infection. IFN-γ production is higher in CCC than ASY patients, and is the most highly expressed cytokine in CCC hearts. Key DTGs that downmodulate IFN-γ, like IL-10, and Ebi3/IL27p28, are higher in ASY patients. Polymorphisms in PRG and DTG are associated with differential disease progression. We thus hypothesize that ASY patients are disease tolerant, while an imbalance of DTG and IFN-γ PRG activity leads to the inflammatory heart damage of CCC.
Collapse
Affiliation(s)
| | - João Paulo Silva Nunes
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Amanda Farage Frade
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Department of Bioengineering, Brazil University, São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Ramendra Pati Pandey
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Marilda Savóia Nascimento
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Jorge Kalil
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| |
Collapse
|
27
|
Pereira NDS, Queiroga TBD, Nunes DF, Andrade CDM, Nascimento MSL, Do-Valle-Matta MA, da Câmara ACJ, Galvão LMDC, Guedes PMM, Chiari E. Innate immune receptors over expression correlate with chronic chagasic cardiomyopathy and digestive damage in patients. PLoS Negl Trop Dis 2018; 12:e0006589. [PMID: 30044791 PMCID: PMC6078325 DOI: 10.1371/journal.pntd.0006589] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 08/06/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022] Open
Abstract
Chronic chagasic cardiomyopathy (CCC) is observed in 30% to 50% of the individuals infected by Trypanosoma cruzi and heart failure is the important cause of death among patients in the chronic phase of Chagas disease. Although some studies have elucidated the role of adaptive immune responses involving T and B lymphocytes in cardiac pathogenesis, the role of innate immunity receptors such as Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in CCC pathophysiology has not yet been determined. In this study, we evaluated the association among innate immune receptors (TLR1-9 and nucleotide-binding domain-like receptor protein 3/NLRP3), its adapter molecules (Myd88, TRIF, ASC and caspase-1) and cytokines (IL-1β, IL-6, IL-12, IL-18, IL-23, TNF-α, and IFN-β) with clinical manifestation, digestive and cardiac function in patients with different clinical forms of chronic Chagas disease. The TLR8 mRNA expression levels were enhanced in the peripheral blood mononuclear cells (PBMC) from digestive and cardiodigestive patients compared to indeterminate and cardiac patients. Furthermore, mRNA expression of IFN-β (cytokine produced after TLR8 activation) was higher in digestive and cardiodigestive patients when compared to indeterminate. Moreover, there was a positive correlation between TLR8 and IFN-β mRNA expression with sigmoid and rectum size. Cardiac and cardiodigestive patients presented higher TLR2, IL-12 and TNF-α mRNA expression than indeterminate and digestive patients. Moreover, cardiac patients also expressed higher levels of NLRP3, ASC and IL-1β mRNAs than indeterminate patients. In addition, we showed a negative correlation among TLR2, IL-1β, IL-12 and TNF-α levels with left ventricular ejection fraction, and positive correlation between NLRP3 with cardiothoracic index, and TLR2, IL-1β and IL-12 with left ventricular mass index. Together, our data suggest that high expression of innate immune receptors in cardiac and digestive patients may induce an enhancement of cytokine expression and participate of cardiac and digestive dysfunction.
Collapse
Affiliation(s)
- Nathalie de Sena Pereira
- Department of Parasitology, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Rio Grande do Norte, Natal, Brazil
- School of Health, Potiguar University, Natal, RN, Brazil
| | | | - Daniela Ferreira Nunes
- Department of Parasitology, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| | - Cléber de Mesquita Andrade
- Department of Biomedical Sciences, University of Rio Grande do Norte State, Rio Grande do Norte, Mossoró, Brazil
| | | | | | | | | | - Paulo Marcos Matta Guedes
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Rio Grande do Norte, Natal, Brazil
| | - Egler Chiari
- Department of Parasitology, Federal University of Minas Gerais, Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
28
|
Rodríguez-Angulo H, Marques J, Mendoza I, Villegas M, Mijares A, Gironès N, Fresno M. Differential cytokine profiling in Chagasic patients according to their arrhythmogenic-status. BMC Infect Dis 2017; 17:221. [PMID: 28327099 PMCID: PMC5361844 DOI: 10.1186/s12879-017-2324-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 03/14/2017] [Indexed: 01/08/2023] Open
Abstract
Background Chagas disease is caused by the protozoan Trypanosoma cruzi and is characterized by heart failure and sudden death. Identifying which factors are involved in evolution and treatment response is actually challenging. Thus, the aim of this work was to determine the Th1/Th17 (IL-6, IL-2, TNF, IL-17 and IFN-γ) and Th2 (IL-4 and IL-10) serum profile in Venezuelan Chagasic patients stratified according amiodarone treatment, hypertension and arrhythmias. Methods Sera from 38 chagasic patients were analyzed to determine the level of cytokines by Multiplexed Bead-Based Immunoassays. ANOVA test was applied to determine differences for each group. Additionally, a Linear Discriminant Analysis (LDA) was applied to observe the accuracy of different cytokines to discriminate between the groups. Results The levels of several cytokines were significantly higher in the high-risk of sudden death and untreated group. LDA showed that IL-2, IFN-γ and IL-10 were the best cytokines for discriminating between high-risk of sudden death and untreated patients versus low-risk of sudden death, treated and control groups. Conclusions High IL-2 levels seem to identify patients with high-risk of sudden death and seems adequate as treatment efficacy marker. To our knowledge, this is the first report about the anti-inflammatory role of the amiodarone in Chagas disease, suggesting an inmunomodulatory effect that may be exploited as coadjutant therapy in chronic Chagas disease. Electronic supplementary material The online version of this article (doi:10.1186/s12879-017-2324-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Juan Marques
- Instituto de Medicina Tropical, Caracas, Venezuela
| | - Ivan Mendoza
- Instituto de Medicina Tropical, Caracas, Venezuela
| | | | - Alfredo Mijares
- Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - Núria Gironès
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049, Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Cantoblanco, 28049, Madrid, Spain
| |
Collapse
|
29
|
Baldissera MD, Sagrillo MR, de Sá MF, Peroza LR, Posser CP, de Brum GF, Peres DS, De Mello ALB, Ferrão M, Souza CF, da Silva AS, Monteiro SG. Increased in cyclooxygenase—2 immunoreactivity and DNA damage in hippocampus of rats infected by Trypanosoma evansi. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s00580-016-2235-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
30
|
Inflammatory and cardiac biomarkers are differentially expressed in clinical stages of Chagas disease. Int J Cardiol 2015; 199:451-9. [PMID: 26277551 DOI: 10.1016/j.ijcard.2015.07.040] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 05/29/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Chagas disease has a long clinically silent period following Trypanosoma cruzi infection and before development of overt clinical pathology; detectable biomarkers of infection and pathogenesis are urgently needed. We tested 22 biomarkers known to be associated with cardiomyopathy to evaluate if a biomarker signature could successfully classify T. cruzi seropositive subjects into clinical Chagas disease stage groups. METHODS This cross-sectional retrospective case-control study enrolled T. cruzi seropositive blood donors (BD) who were further characterized as having chronic Chagas cardiomyopathy (CC-BD) or not (nonCC-BD) and seronegative (SN) control donors; we also included clinically diagnosed Chagas cardiomyopathy patients (CC-P). All subjects underwent a health history questionnaire, medical examination, electro- and echocardiograms (ECG and Echo) and phlebotomy. Biomarkers were measured on blinded samples by luminex bead array and Ortho VITROS. RESULTS A clear biomarker pattern was observed only in more severe cardiac disease; this pattern included significantly elevated levels of inflammatory cytokines IFN-γ, IL-6, IL-10 and TNF-α and soluble cardiovascular disease biomarkers CK-MB, troponin, myoglobin, VCAM and NTproBNP while there were lower levels of MPO, PAI-1, and MCP-1. The markers determined to be the most predictive of disease by ROC curve analysis were NTproBNP and T. cruzi PCR status. CONCLUSIONS Although many biomarkers demonstrated increased or decreased concentrations among the clinical forms of Chagas disease, NTproBNP and T. cruzi PCR were the only tests that would independently be of clinical value for disease staging, in concert with ECG, Echo and clinical assessments.
Collapse
|
31
|
Nogueira LG, Frade AF, Ianni BM, Laugier L, Pissetti CW, Cabantous S, Baron M, de Lima Peixoto G, de Melo Borges A, Donadi E, Marin-Neto JA, Schmidt A, Dias F, Saba B, Wang HTL, Fragata A, Sampaio M, Hirata MH, Buck P, Mady C, Martinelli M, Lensi M, Siqueira SF, Pereira AC, Rodrigues V, Kalil J, Chevillard C, Cunha-Neto E. Functional IL18 polymorphism and susceptibility to Chronic Chagas Disease. Cytokine 2015; 73:79-83. [DOI: 10.1016/j.cyto.2015.01.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 01/28/2015] [Accepted: 01/29/2015] [Indexed: 01/09/2023]
|
32
|
Pentoxifylline reverses chronic experimental Chagasic cardiomyopathy in association with repositioning of abnormal CD8+ T-cell response. PLoS Negl Trop Dis 2015; 9:e0003659. [PMID: 25789471 PMCID: PMC4366205 DOI: 10.1371/journal.pntd.0003659] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 02/27/2015] [Indexed: 12/22/2022] Open
Abstract
Background Chronic chagasic cardiomyopathy (CCC), the main clinical sign of Chagas disease, is associated with systemic CD8+ T-cell abnormalities and CD8-enriched myocarditis occurring in an inflammatory milieu. Pentoxifylline (PTX), a phosphodiesterase inhibitor, has immunoregulatory and cardioprotective properties. Here, we tested PTX effects on CD8+ T-cell abnormalities and cardiac alterations using a model of experimental Chagas’ heart disease. Methodology/Principal Findings C57BL/6 mice chronically infected by the Colombian Trypanosoma cruzi strain and presenting signs of CCC were treated with PTX. The downmodulation of T-cell receptors on CD8+ cells induced by T. cruzi infection was rescued by PTX therapy. Also, PTX reduced the frequency of CD8+ T-cells expressing activation and migration markers in the spleen and the activation of blood vessel endothelial cells and the intensity of inflammation in the heart tissue. Although preserved interferon-gamma production systemically and in the cardiac tissue, PTX therapy reduced the number of perforin+ cells invading this tissue. PTX did not alter parasite load, but hampered the progression of heart injury, improving connexin 43 expression and decreasing fibronectin overdeposition. Further, PTX reversed electrical abnormalities as bradycardia and prolonged PR, QTc and QRS intervals in chronically infected mice. Moreover, PTX therapy improved heart remodeling since reduced left ventricular (LV) hypertrophy and restored the decreased LV ejection fraction. Conclusions/Significance PTX therapy ameliorates critical aspects of CCC and repositioned CD8+ T-cell response towards homeostasis, reinforcing that immunological abnormalities are crucially linked, as cause or effect, to CCC. Therefore, PTX emerges as a candidate to treat the non-beneficial immune deregulation associated with chronic Chagas' heart disease and to improve prognosis. Chronic chagasic cardiomyopathy (CCC) is the main clinical manifestation of Chagas disease (CD), a neglected illness caused by the protozoan parasite Trypanosoma cruzi. More than hundred years after its discovery, CD continues to be a public health problem and millions of chronically infected people wait for an effective treatment. Chagasic cardiomyopathy is associated with CD8+ T-cell-enriched myocarditis, fibrosis and cardiac electrical and structural abnormalities, frequently progressing to heart failure. Presently, the available therapies only mitigate symptoms of CCC. Abnormalities in CD8+ T-cell compartment are present in CCC patients. Recently, we described the importance of CD8+ T-cells in the pathogenesis of CCC. Therefore, our proposal was to interfere with abnormalities of CD8+ T-cells glimpsing a better prognosis for CCC. Using PTX, an affordable drug with immunomodulatory properties on T-cells and cardioprotective effects in non-infections disease, we bring a therapeutic candidate for treating CCC. PTX therapy downmodulated detrimental CD8+ T-cells and promoted T. cruzi-specific interferon-gamma-producing T-cells. Importantly, chronic chagasic electrical and echocardiographic alterations were reversed by PTX therapy. Future studies may test the use of PTX combined with trypanocidal drug or as a vaccine adjuvant to improve the quality of life of chronic CD patients.
Collapse
|
33
|
Ferreira LRP, Frade AF, Baron MA, Navarro IC, Kalil J, Chevillard C, Cunha-Neto E. Interferon-γ and other inflammatory mediators in cardiomyocyte signaling during Chagas disease cardiomyopathy. World J Cardiol 2014; 6:782-790. [PMID: 25228957 PMCID: PMC4163707 DOI: 10.4330/wjc.v6.i8.782] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/29/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
Chagas disease cardiomyopathy (CCC), the main consequence of Trypanosoma cruzi (T.cruzi) infection, is an inflammatory cardiomyopathy that develops in up to 30% of infected individuals. The heart inflammation in CCC patients is characterized by a Th1 T cell-rich myocarditis with increased production of interferon (IFN)-γ, produced by the CCC myocardial infiltrate and detected at high levels in the periphery. IFN-γ has a central role in the cardiomyocyte signaling during both acute and chronic phases of T.cruzi infection. In this review, we have chosen to focus in its pleiotropic mode of action during CCC, which may ultimately be the strongest driver towards pathological remodeling and heart failure. We describe here the antiparasitic protective and pathogenic dual role of IFN-γ in Chagas disease.
Collapse
|
34
|
Cunha-Neto E, Chevillard C. Chagas disease cardiomyopathy: immunopathology and genetics. Mediators Inflamm 2014; 2014:683230. [PMID: 25210230 PMCID: PMC4152981 DOI: 10.1155/2014/683230] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/05/2014] [Accepted: 08/05/2014] [Indexed: 02/06/2023] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, is endemic in Latin America and affects ca. 10 million people worldwide. About 30% of Chagas disease patients develop chronic Chagas disease cardiomyopathy (CCC), a particularly lethal inflammatory cardiomyopathy that occurs decades after the initial infection, while most patients remain asymptomatic. Mortality rate is higher than that of noninflammatory cardiomyopathy. CCC heart lesions present a Th1 T-cell-rich myocarditis, with cardiomyocyte hypertrophy and prominent fibrosis. Data suggest that the myocarditis plays a major pathogenetic role in disease progression. Major unmet goals include the thorough understanding of disease pathogenesis and therapeutic targets and identification of prognostic genetic factors. Chagas disease thus remains a neglected disease, with no vaccines or antiparasitic drugs proven efficient in chronically infected adults, when most patients are diagnosed. Both familial aggregation of CCC cases and the fact that only 30% of infected patients develop CCC suggest there might be a genetic component to disease susceptibility. Moreover, previous case-control studies have identified some genes associated to human susceptibility to CCC. In this paper, we will review the immunopathogenesis and genetics of Chagas disease, highlighting studies that shed light on the differential progression of Chagas disease patients to CCC.
Collapse
Affiliation(s)
- Edecio Cunha-Neto
- Heart Institute (InCor), University of São Paulo School of Medicine, Avenida Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9° Andar, 05406-000 São Paulo, SP, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, 05406-000 São Paulo, SP, Brazil
| | | |
Collapse
|
35
|
Myocardial gene expression of T-bet, GATA-3, Ror-γt, FoxP3, and hallmark cytokines in chronic Chagas disease cardiomyopathy: an essentially unopposed TH1-type response. Mediators Inflamm 2014; 2014:914326. [PMID: 25152568 PMCID: PMC4134835 DOI: 10.1155/2014/914326] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/08/2014] [Indexed: 11/17/2022] Open
Abstract
Background. Chronic Chagas disease cardiomyopathy (CCC), a late consequence of Trypanosoma cruzi infection, is an inflammatory cardiomyopathy with prognosis worse than those of noninflammatory etiology (NIC). Although the T cell-rich myocarditis is known to play a pathogenetic role, the relative contribution of each of the functional T cell subsets has never been thoroughly investigated. We therefore assessed gene expression of cytokines and transcription factors involved in differentiation and effector function of each functional T cell subset (TH1/TH2/TH17/Treg) in CCC, NIC, and heart donor myocardial samples. Methods and Results. Quantitative PCR showed markedly upregulated expression of IFN-γ and transcription factor T-bet, and minor increases of GATA-3; FoxP3 and CTLA-4; IL-17 and IL-18 in CCC as compared with NIC samples. Conversely, cytokines expressed by TH2 cells (IL-4, IL-5, and IL-13) or associated with Treg (TGF-β
and IL-10) were not upregulated in CCC myocardium. Expression of TH1-related genes such as T-bet, IFN-γ, and IL-18 correlated with ventricular dilation, FoxP3, and CTLA-4. Conclusions. Results are consistent with a strong local TH1-mediated response in most samples, possibly associated with pathological myocardial remodeling, and a proportionally smaller FoxP3+CTLA4+ Treg cell population, which is unable to completely curb IFN-γ production in CCC myocardium, therefore fueling inflammation.
Collapse
|
36
|
Induction of IL-12 production in human peripheral monocytes by Trypanosoma cruzi Is mediated by glycosylphosphatidylinositol-anchored mucin-like glycoproteins and potentiated by IFN- γ and CD40-CD40L interactions. Mediators Inflamm 2014; 2014:345659. [PMID: 25120285 PMCID: PMC4120781 DOI: 10.1155/2014/345659] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/16/2014] [Indexed: 12/04/2022] Open
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi (T. cruzi), is characterized by immunopathology driven by IFN-γ secreting Th1-like T cells. T. cruzi has a thick coat of mucin-like glycoproteins covering its surface, which plays an important role in parasite invasion and host immunomodulation. It has been extensively described that T. cruzi or its products—like GPI anchors isolated from GPI-anchored mucins from the trypomastigote life cycle stage (tGPI-mucins)—are potent inducers of proinflammatory responses (i.e., cytokines and NO production) by IFN-γ primed murine macrophages. However, little is known about whether T. cruzi or GPI-mucins exert a similar action in human cells. We therefore decided to further investigate the in vitro cytokine production profile from human mononuclear cells from uninfected donors exposed to T. cruzi as well as tGPI-mucins. We observed that both living T. cruzi trypomastigotes and tGPI-mucins are potent inducers of IL-12 by human peripheral blood monocytes and this effect depends on CD40-CD40L interaction and IFN-γ. Our findings suggest that the polarized T1-type cytokine profile seen in T. cruzi infected patients might be a long-term effect of IL-12 production induced by lifelong exposure to T. cruzi tGPI-mucins.
Collapse
|
37
|
Chagas disease: still many unsolved issues. Mediators Inflamm 2014; 2014:912965. [PMID: 25104883 PMCID: PMC4101227 DOI: 10.1155/2014/912965] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/15/2014] [Indexed: 11/18/2022] Open
Abstract
Over the past 20 years, the immune effector mechanisms involved in the control of Trypanosoma cruzi, as well as the receptors participating in parasite recognition by cells of the innate immune system, have been largely described. However, the main questions on the physiopathology of Chagas disease remain unanswered: “Why does the host immune system fail to provide sterile immunity?” and “Why do only a proportion of infected individuals develop chronic pathology?” In this review, we describe the mechanisms proposed to explain the inability of the immune system to eradicate the parasite and the elements that allow the development of chronic heart disease. Moreover, we discuss the possibility that the inability of infected cardiomyocytes to sense intracellular T. cruzi contributes to parasite persistence in the heart and the development of chronic pathology.
Collapse
|
38
|
Plasma cytokine expression is associated with cardiac morbidity in chagas disease. PLoS One 2014; 9:e87082. [PMID: 24603474 PMCID: PMC3945957 DOI: 10.1371/journal.pone.0087082] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 12/18/2013] [Indexed: 01/01/2023] Open
Abstract
The expression of immune response appears to be associated with morbidity in Chagas disease. However, the studies in this field have usually employed small samples of patients and statistical analyses that do not consider the wide dispersion of cytokine production observed in these patients. The aim of this study was to evaluate the plasma cytokine levels in well-defined clinical polar groups of chagasic patients divided into categories that better reflect the wide cytokine profile and its relationship with morbidity. Patients infected with Trypanosoma cruzi (T. cruzi) were grouped as indeterminate (IND) and cardiac (CARD) forms ranging from 23 to 69 years of age (mean of 45.6±11.25). The IND group included 82 individuals, ranging from 24 to 66 years of age (mean of 39.6±10.3). The CARD group included 94 patients ranging from 23 to 69 years of age (mean of 48±12.52) presenting dilated cardiomyopathy. None of the patients have undergone chemotherapeutic treatment, nor had been previously treated for T. cruzi infection. Healthy non-chagasic individuals, ranging from 29 to 55 years of age (mean of 42.6±8.8) were included as a control group (NI). IND patients have a higher intensity of interleukin 10 (IL-10) expression when compared with individuals in the other groups. By contrast, inflammatory cytokine expression, such as interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and interleukin 1 beta (IL-1β), proved to be the highest in the CARD group. Correlation analysis showed that higher IL-10 expression was associated with better cardiac function, as determined by left ventricular ejection fraction and left ventricular diastolic diameter values. Altogether, these findings reinforce the concept that a fine balance between regulatory and inflammatory cytokines represents a key element in the establishment of distinct forms of chronic Chagas disease.
Collapse
|
39
|
Frade AF, Pissetti CW, Ianni BM, Saba B, Lin-Wang HT, Nogueira LG, de Melo Borges A, Buck P, Dias F, Baron M, Ferreira LRP, Schmidt A, Marin-Neto JA, Hirata M, Sampaio M, Fragata A, Pereira AC, Donadi E, Kalil J, Rodrigues V, Cunha-Neto E, Chevillard C. Genetic susceptibility to Chagas disease cardiomyopathy: involvement of several genes of the innate immunity and chemokine-dependent migration pathways. BMC Infect Dis 2013; 13:587. [PMID: 24330528 PMCID: PMC3866603 DOI: 10.1186/1471-2334-13-587] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 12/04/2013] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Chagas disease, caused by the protozoan Trypanosoma cruzi is endemic in Latin America. Thirty percent of infected individuals develop chronic Chagas cardiomyopathy (CCC), an inflammatory dilated cardiomyopathy that is, by far, the most important clinical consequence of T. cruzi infection. The others remain asymptomatic (ASY). A possible genetic component to disease progression was suggested by familial aggregation of cases and the association of markers of innate and adaptive immunity genes with CCC development. Migration of Th1-type T cells play a major role in myocardial damage. METHODS Our genetic analysis focused on CCR5, CCL2 and MAL/TIRAP genes. We used the Tag SNPs based approach, defined to catch all the genetic information from each gene. The study was conducted on a large Brazilian population including 315 CCC cases and 118 ASY subjects. RESULTS The CCL2rs2530797A/A and TIRAPrs8177376A/A were associated to an increase susceptibility whereas the CCR5rs3176763C/C genotype is associated to protection to CCC. These associations were confirmed when we restricted the analysis to severe CCC, characterized by a left ventricular ejection fraction under 40%. CONCLUSIONS Our data show that polymorphisms affecting key molecules involved in several immune parameters (innate immunity signal transduction and T cell/monocyte migration) play a role in genetic susceptibility to CCC development. This also points out to the multigenic character of CCC, each polymorphism imparting a small contribution. The identification of genetic markers for CCC will provide information for pathogenesis as well as therapeutic targets.
Collapse
Affiliation(s)
- Amanda Farage Frade
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
- Aix-Marseille Université, INSERM, GIMP UMR_S906, Faculté de médecine, 27 bd Jean Moulin, Marseille, cedex 05 13385, France
| | - Cristina Wide Pissetti
- Laboratory of Immunology, Universidade Federal do Triângulo Mineiro (UFTM), 40 Frei Paulino, Uberaba, MG 48036-180, Brazil
| | - Barbara Maria Ianni
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
| | - Bruno Saba
- Instituto de Cardiologia Dante Pazzanese (IDPC), Avenida Dante Pazzanese 500 - Ibirapuera, Sâo Paulo, SP 04012-909, Brazil
| | - Hui Tzu Lin-Wang
- Instituto de Cardiologia Dante Pazzanese (IDPC), Avenida Dante Pazzanese 500 - Ibirapuera, Sâo Paulo, SP 04012-909, Brazil
| | - Luciana Gabriel Nogueira
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
| | - Ariana de Melo Borges
- Laboratory of Immunology, Universidade Federal do Triângulo Mineiro (UFTM), 40 Frei Paulino, Uberaba, MG 48036-180, Brazil
| | - Paula Buck
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
| | - Fabrício Dias
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Av. Bandeirantes, 4900 - Monte Alegre 15059-900, Ribeirão Preto, SP, Brazil
| | - Monique Baron
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
| | - Ludmila Rodrigues Pinto Ferreira
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
| | - Andre Schmidt
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Av. Bandeirantes, 4900 - Monte Alegre 15059-900, Ribeirão Preto, SP, Brazil
| | - José Antonio Marin-Neto
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Av. Bandeirantes, 4900 - Monte Alegre 15059-900, Ribeirão Preto, SP, Brazil
| | - Mario Hirata
- Instituto de Cardiologia Dante Pazzanese (IDPC), Avenida Dante Pazzanese 500 - Ibirapuera, Sâo Paulo, SP 04012-909, Brazil
| | - Marcelo Sampaio
- Instituto de Cardiologia Dante Pazzanese (IDPC), Avenida Dante Pazzanese 500 - Ibirapuera, Sâo Paulo, SP 04012-909, Brazil
| | - Abílio Fragata
- Instituto de Cardiologia Dante Pazzanese (IDPC), Avenida Dante Pazzanese 500 - Ibirapuera, Sâo Paulo, SP 04012-909, Brazil
| | - Alexandre Costa Pereira
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
| | - Eduardo Donadi
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Av. Bandeirantes, 4900 - Monte Alegre 15059-900, Ribeirão Preto, SP, Brazil
| | - Jorge Kalil
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, SP 06504-000, Brazil
| | - Virmondes Rodrigues
- Laboratory of Immunology, Universidade Federal do Triângulo Mineiro (UFTM), 40 Frei Paulino, Uberaba, MG 48036-180, Brazil
| | - Edecio Cunha-Neto
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), Av. Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9º andar, São Paulo, SP 06504-000, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, SP 06504-000, Brazil
| | - Christophe Chevillard
- Aix-Marseille Université, INSERM, GIMP UMR_S906, Faculté de médecine, 27 bd Jean Moulin, Marseille, cedex 05 13385, France
| |
Collapse
|
40
|
Oliveira LGR, Kuehn CC, dos Santos CD, Miranda MA, da Costa CMB, Mendonça VJ, do Prado JC. Protective actions of melatonin against heart damage during chronic Chagas disease. Acta Trop 2013; 128:652-8. [PMID: 24055715 DOI: 10.1016/j.actatropica.2013.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 09/06/2013] [Accepted: 09/10/2013] [Indexed: 01/05/2023]
Abstract
Chronic cardiomyopathy is the most important clinical form of Chagas disease, and it is characterised by myocarditis that is associated with fibrosis and organ dysfunction. Alternative treatment options are important tools to modulate host immune responses. The main goal of this work was to evaluate the anti-inflammatory actions of melatonin during the chronic phase of Chagas disease. TNF-α, IL-10 and nitrite concentrations were evaluated as predictive factors of immune modulation. Creatine phosphokinase-MB (CK-MB), cardiac inflammatory foci and heart weight were assessed to evaluate the efficacy of the melatonin treatment. Male Wistar rats were infected with 1×10(5) blood trypomastigotes of the Y strain of Trypanosoma cruzi and kept untreated for 60 days to mimic chronic infection. After this period, the rats were orally treated with melatonin 50mg/kg/day, and the experiments were performed 90, 120, and 180 days post-infection. Melatonin treatment significantly increased the concentration of IL-10 and reduced the concentrations of NO and TNF-α produced by cardiomyocytes. Furthermore, it led to decreased heart weight, serum CK-MB levels and inflammatory foci when compared to the untreated and infected control groups. We conclude that melatonin therapy is effective at protecting animals against the harmful cardiac inflammatory response that is characteristic of chronic T. cruzi infection.
Collapse
|
41
|
Nunes DF, Guedes PMDM, de Mesquita Andrade C, Câmara ACJD, Chiari E, Galvão LMDC. Troponin T autoantibodies correlate with chronic cardiomyopathy in human Chagas disease. Trop Med Int Health 2013; 18:1180-92. [DOI: 10.1111/tmi.12169] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Daniela Ferreira Nunes
- Graduate Program in Health Sciences; Center for Health Sciences; Federal University of Rio Grande do Norte; Natal; Brazil
| | - Paulo Marcos da Matta Guedes
- Department of Microbiology and Parasitology; Center for Biosciences; Federal University of Rio Grande do Norte; Natal; Brazil
| | - Cléber de Mesquita Andrade
- Graduate Program in Health Sciences; Center for Health Sciences; Federal University of Rio Grande do Norte; Natal; Brazil
| | - Antonia Cláudia Jácome da Câmara
- Department of Clinical and Toxicological Analyses; Center for Health Sciences; Federal University of Rio Grande do Norte; Natal; Brazil
| | - Egler Chiari
- Department of Parasitology; Institute of Biological Sciences; Federal University of Minas Gerais; Belo Horizonte; Brazil
| | - Lúcia Maria da Cunha Galvão
- Graduate Program in Health Sciences; Center for Health Sciences; Federal University of Rio Grande do Norte; Natal; Brazil
| |
Collapse
|
42
|
de Meis J, Barreto de Albuquerque J, Silva Dos Santos D, Farias-de-Oliveira DA, Berbert LR, Cotta-de-Almeida V, Savino W. Trypanosoma cruzi Entrance through Systemic or Mucosal Infection Sites Differentially Modulates Regional Immune Response Following Acute Infection in Mice. Front Immunol 2013; 4:216. [PMID: 23898334 PMCID: PMC3724200 DOI: 10.3389/fimmu.2013.00216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/13/2013] [Indexed: 12/11/2022] Open
Abstract
Acute Chagas disease is characterized by a systemic infection that leads to the strong activation of the adaptive immune response. Outbreaks of oral contamination by the infective protozoan Trypanosoma cruzi are frequent in Brazil and other Latin American countries, and an increased severity of clinical manifestations and mortality is observed in infected patients. These findings have elicited questions about the specific responses triggered after T. cruzi entry via mucosal sites, possibly modulating local immune mechanisms, and further impacting regional and systemic immunity. Here, we provide evidence for the existence of differential lymphoid organ responses in experimental models of acute T. cruzi infection.
Collapse
Affiliation(s)
- Juliana de Meis
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | | | | | | | | | | | | |
Collapse
|
43
|
Nogueira LG, Santos RHB, Ianni BM, Fiorelli AI, Mairena EC, Benvenuti LA, Frade A, Donadi E, Dias F, Saba B, Wang HTL, Fragata A, Sampaio M, Hirata MH, Buck P, Mady C, Bocchi EA, Stolf NA, Kalil J, Cunha-Neto E. Myocardial chemokine expression and intensity of myocarditis in Chagas cardiomyopathy are controlled by polymorphisms in CXCL9 and CXCL10. PLoS Negl Trop Dis 2012; 6:e1867. [PMID: 23150742 PMCID: PMC3493616 DOI: 10.1371/journal.pntd.0001867] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 09/03/2012] [Indexed: 01/24/2023] Open
Abstract
Background Chronic Chagas cardiomyopathy (CCC), a life-threatening inflammatory dilated cardiomyopathy, affects 30% of the approximately 8 million patients infected by Trypanosoma cruzi. Even though the Th1 T cell-rich myocarditis plays a pivotal role in CCC pathogenesis, little is known about the factors controlling inflammatory cell migration to CCC myocardium. Methods and Results Using confocal immunofluorescence and quantitative PCR, we studied cell surface staining and gene expression of the CXCR3, CCR4, CCR5, CCR7, CCR8 receptors and their chemokine ligands in myocardial samples from end-stage CCC patients. CCR5+, CXCR3+, CCR4+, CCL5+ and CXCL9+ mononuclear cells were observed in CCC myocardium. mRNA expression of the chemokines CCL5, CXCL9, CXCL10, CCL17, CCL19 and their receptors was upregulated in CCC myocardium. CXCL9 mRNA expression directly correlated with the intensity of myocarditis, as well as with mRNA expression of CXCR3, CCR4, CCR5, CCR7, CCR8 and their ligands. We also analyzed single-nucleotide polymorphisms for genes encoding the most highly expressed chemokines and receptors in a cohort of Chagas disease patients. CCC patients with ventricular dysfunction displayed reduced genotypic frequencies of CXCL9 rs10336 CC, CXCL10 rs3921 GG, and increased CCR5 rs1799988CC as compared to those without dysfunction. Significantly, myocardial samples from CCC patients carrying the CXCL9/CXCL10 genotypes associated to a lower risk displayed a 2–6 fold reduction in mRNA expression of CXCL9, CXCL10, and other chemokines and receptors, along with reduced intensity of myocarditis, as compared to those with other CXCL9/CXCL10 genotypes. Conclusions Results may indicate that genotypes associated to reduced risk in closely linked CXCL9 and CXCL10 genes may modulate local expression of the chemokines themselves, and simultaneously affect myocardial expression of other key chemokines as well as intensity of myocarditis. Taken together our results may suggest that CXCL9 and CXCL10 are master regulators of myocardial inflammatory cell migration, perhaps affecting clinical progression to the life-threatening form of CCC. Chronic Chagas cardiomyopathy (CCC) is an inflammatory heart disease that affects millions in Latin America, and in growing numbers in USA and Europe. Survival among CCC patients is shorter than among patients with cardiomyopathy of non-inflammatory etiology. This suggests that the inflammatory cell influx plays an important pathogenic role in CCC. However, little is known about the factors that maintain this myocardial inflammation. We hypothesized that Th1 T cell-attracting chemokines, involved in driving leukocyte migration, could play a role in myocardial inflammation. Herein, we have analyzed expression of several chemokines and receptors in heart tissue from patients with CCC and controls. We found inflammatory cells expressing chemokines and receptors consistent with Th1 T cell influx into CCC myocardium. mRNA expression levels of the chemokine CXCL9 correlated with inflammation. We also studied whether genetic variations in these genes could be associated to CCC development. Polymorphisms in CXCL9, CXCL10 and CCR5 were associated to differential risk of progression to the more severe form of CCC. Polymorphisms of CXCL9 and CXCL10 were also associated to the intensity of myocardial inflammation and chemokine expression. These results suggest that such chemokines may be master regulators of myocardial inflammatory cell migration, perhaps affecting clinical progression to severe CCC.
Collapse
Affiliation(s)
- Luciana Gabriel Nogueira
- Laboratory of Immunology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | | | - Barbara Maria Ianni
- Myocardiopathies Unit, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Alfredo Inácio Fiorelli
- Divison of Surgery, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Eliane Conti Mairena
- Laboratory of Immunology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Luiz Alberto Benvenuti
- Divison of Pathology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Amanda Frade
- Laboratory of Immunology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Eduardo Donadi
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, São Paulo, Brazil
| | - Fabrício Dias
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, São Paulo, Brazil
| | - Bruno Saba
- Dante Pazzanese Institute of Cardiology and Heart Failure Unit, São Paulo, Brazil
| | - Hui-Tzu Lin Wang
- Dante Pazzanese Institute of Cardiology and Heart Failure Unit, São Paulo, Brazil
| | - Abilio Fragata
- Dante Pazzanese Institute of Cardiology and Heart Failure Unit, São Paulo, Brazil
| | - Marcelo Sampaio
- Dante Pazzanese Institute of Cardiology and Heart Failure Unit, São Paulo, Brazil
| | | | - Paula Buck
- Myocardiopathies Unit, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Charles Mady
- Myocardiopathies Unit, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Edimar Alcides Bocchi
- Transplantation and Heart Failure Unit, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Noedir Antonio Stolf
- Divison of Surgery, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor), School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
44
|
Foxp3+CD25high CD4+ regulatory T cells from indeterminate patients with Chagas disease can suppress the effector cells and cytokines and reveal altered correlations with disease severity. Immunobiology 2012; 217:768-77. [DOI: 10.1016/j.imbio.2012.04.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 03/26/2012] [Accepted: 04/27/2012] [Indexed: 12/17/2022]
|
45
|
Teixeira PC, Frade AF, Nogueira LG, Kalil J, Chevillard C, Cunha-Neto E. Pathogenesis of Chagas disease cardiomyopathy. World J Clin Infect Dis 2012; 2:39-53. [DOI: 10.5495/wjcid.v2.i3.39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chagas disease, or American trypanosomiasis, is a parasitic infection caused by the flagellate protozoan Trypanosoma cruzi. Chagas disease is mainly affecting rural populations in Mexico and Central and South America. The World Health Organization estimates that 300 000 new cases of Chagas disease occur every year and approximately 20 000 deaths are attributable to Chagas. However, this organisation classified Chagas disease as a neglected tropical disease. The economic burden of this disease is significant. In many Latin American countries, the direct and indirect costs, including the cost of health care in dollars and loss of productivity, attributable to Chagas disease ranges from $40 million to in excess of $800 million per nation per annum. So, it remains a contemporary public health concern. In chronic phase, mortality is primarily due to the rhythm disturbances and congestive heart failure that result from the chronic inflammatory cardiomyopathy (CCC) due to the persistence presence of parasites in the heart tissue. Mechanisms underlying differential progression to CCC are still incompletely understood. In the last decades immunological proteomic genetic approaches lead to significant results which help to disperse the veil covering the knowledge of the pathogenic process. Here, we reported these significant progresses.
Collapse
|
46
|
Silverio JC, Pereira IR, Cipitelli MDC, Vinagre NF, Rodrigues MM, Gazzinelli RT, Lannes-Vieira J. CD8+ T-cells expressing interferon gamma or perforin play antagonistic roles in heart injury in experimental Trypanosoma cruzi-elicited cardiomyopathy. PLoS Pathog 2012; 8:e1002645. [PMID: 22532799 PMCID: PMC3330123 DOI: 10.1371/journal.ppat.1002645] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 03/01/2012] [Indexed: 12/12/2022] Open
Abstract
In Chagas disease, CD8(+) T-cells are critical for the control of Trypanosoma cruzi during acute infection. Conversely, CD8(+) T-cell accumulation in the myocardium during chronic infection may cause tissue injury leading to chronic chagasic cardiomyopathy (CCC). Here we explored the role of CD8(+) T-cells in T. cruzi-elicited heart injury in C57BL/6 mice infected with the Colombian strain. Cardiomyocyte lesion evaluated by creatine kinase-MB isoenzyme activity levels in the serum and electrical abnormalities revealed by electrocardiogram were not associated with the intensity of heart parasitism and myocarditis in the chronic infection. Further, there was no association between heart injury and systemic anti-T. cruzi CD8(+) T-cell capacity to produce interferon-gamma (IFNγ) and to perform specific cytotoxicity. Heart injury, however, paralleled accumulation of anti-T. cruzi cells in the cardiac tissue. In T. cruzi infection, most of the CD8(+) T-cells segregated into IFNγ(+) perforin (Pfn)(neg) or IFNγ(neg)Pfn(+) cell populations. Colonization of the cardiac tissue by anti-T. cruzi CD8(+)Pfn(+) cells paralleled the worsening of CCC. The adoptive cell transfer to T. cruzi-infected cd8(-/-) recipients showed that the CD8(+) cells from infected ifnγ(-/-)pfn(+/+) donors migrate towards the cardiac tissue to a greater extent and caused a more severe cardiomyocyte lesion than CD8(+) cells from ifnγ(+/+)pfn(-/-) donors. Moreover, the reconstitution of naïve cd8(-/-) mice with CD8(+) cells from naïve ifnγ(+/+)pfn(-/-) donors ameliorated T. cruzi-elicited heart injury paralleled IFNγ(+) cells accumulation, whereas reconstitution with CD8(+) cells from naïve ifnγ(-/-)pfn(+/+) donors led to an aggravation of the cardiomyocyte lesion, which was associated with the accumulation of Pfn(+) cells in the cardiac tissue. Our data support a possible antagonist effect of CD8(+)Pfn(+) and CD8(+)IFNγ(+) cells during CCC. CD8(+)IFNγ(+) cells may exert a beneficial role, whereas CD8(+)Pfn(+) may play a detrimental role in T. cruzi-elicited heart injury.
Collapse
Affiliation(s)
| | - Isabela Resende Pereira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | | | | | | | - Ricardo Tostes Gazzinelli
- Laboratório de Imunoparasitologia, Instituto Rene Rachou, Fiocruz, Minas Gerais, Brazil
- Departamento de Imunologia e Bioquímica, ICB, UFMG, Minas Gerais, Brazil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
47
|
Different infective forms trigger distinct immune response in experimental Chagas disease. PLoS One 2012; 7:e32912. [PMID: 22412949 PMCID: PMC3296760 DOI: 10.1371/journal.pone.0032912] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 02/04/2012] [Indexed: 11/26/2022] Open
Abstract
Although metacyclic and blood trypomastigotes are completely functional in relation to parasite-host interaction and/or target cell invasion, they differ in the molecules present on the surface. Thus, aspects related to the variability that the forms of T. cruzi interacts with host cells may lead to fundamental implications on the immune response against this parasite and, consequently, the clinical evolution of Chagas disease. We have shown that BT infected mice presented higher levels of parasitemia during all the acute phase of infection. Moreover, the infection with either MT or BT forms resulted in increased levels of total leukocytes, monocytes and lymphocytes, specifically later for MT and earlier for BT. The infection with BT forms presented earlier production of proinflammatory cytokine TNF-α and later of IFN-γ by both T cells subpopulations. This event was accompanied by an early cardiac inflammation with an exacerbation of this process at the end of the acute phase. On the other hand, infection with MT forms result in an early production of IFN-γ, with subsequent control in the production of this cytokine by IL-10, which provided to these animals an immunomodulatory profile in the end of the acute phase. These results are in agreement with what was found for cardiac inflammation where animals infected with MT forms showed intense cardiac inflammation later at infection, with a decrease in the same at the end of this phase. In summary, our findings emphasize the importance of taking into account the inoculums source of T. cruzi, since vectorial or transfusional routes of T. cruzi infection may trigger distinct parasite-host interactions during the acute phase that may influence relevant biological aspects of chronic Chagas disease.
Collapse
|
48
|
Roffê E, Rothfuchs AG, Santiago HC, Marino APMP, Ribeiro-Gomes FL, Eckhaus M, Antonelli LRV, Murphy PM. IL-10 limits parasite burden and protects against fatal myocarditis in a mouse model of Trypanosoma cruzi infection. THE JOURNAL OF IMMUNOLOGY 2011; 188:649-60. [PMID: 22156594 DOI: 10.4049/jimmunol.1003845] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chagas' disease is a zoonosis prevalent in Latin America that is caused by the protozoan Trypanosoma cruzi. The immunopathogenesis of cardiomyopathy, the main clinical problem in Chagas' disease, has been extensively studied but is still poorly understood. In this study, we systematically compared clinical, microbiologic, pathologic, immunologic, and molecular parameters in two mouse models with opposite susceptibility to acute myocarditis caused by the myotropic Colombiana strain of T. cruzi: C3H/HeSnJ (100% mortality, uncontrolled parasitism) and C57BL/6J (<10% mortality, controlled parasitism). T. cruzi induced differential polarization of immunoregulatory cytokine mRNA expression in the hearts of C57BL/6J versus C3H/HeSnJ mice; however, most differences were small. The difference in IL-10 expression was exceptional (C57BL/6J 8.7-fold greater than C3H/HeSnJ). Consistent with this, hearts from infected C57BL/6J mice, but not C3H/HeSnJ mice, had a high frequency of total IL-10-producing CD8(+) T cells and both CD4(+) and CD8(+) subsets of IFN-γ(+)IL-10(+) double-producing T cells. Furthermore, T. cruzi infection of IL-10(-/-) C57BL/6J mice phenocopied fatal infection in wild-type C3H/HeSnJ mice with complete loss of parasite control. Adoptive transfer experiments indicated that T cells were a source of protective IL-10. Thus, in this system, IL-10 production by T cells promotes T. cruzi control and protection from fatal acute myocarditis.
Collapse
Affiliation(s)
- Ester Roffê
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Teixeira PC, Santos RHB, Fiorelli AI, Bilate AMB, Benvenuti LA, Stolf NA, Kalil J, Cunha-Neto E. Selective decrease of components of the creatine kinase system and ATP synthase complex in chronic Chagas disease cardiomyopathy. PLoS Negl Trop Dis 2011; 5:e1205. [PMID: 21738806 PMCID: PMC3125151 DOI: 10.1371/journal.pntd.0001205] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 05/01/2011] [Indexed: 01/25/2023] Open
Abstract
Background Chronic Chagas disease cardiomyopathy (CCC) is an inflammatory dilated cardiomyopathy with a worse prognosis than other cardiomyopathies. CCC occurs in 30 % of individuals infected with Trypanosoma cruzi, endemic in Latin America. Heart failure is associated with impaired energy metabolism, which may be correlated to contractile dysfunction. We thus analyzed the myocardial gene and protein expression, as well as activity, of key mitochondrial enzymes related to ATP production, in myocardial samples of end-stage CCC, idiopathic dilated (IDC) and ischemic (IC) cardiomyopathies. Methodology/Principal Findings Myocardium homogenates from CCC (N = 5), IC (N = 5) and IDC (N = 5) patients, as well as from heart donors (N = 5) were analyzed for protein and mRNA expression of mitochondrial creatine kinase (CKMit) and muscular creatine kinase (CKM) and ATP synthase subunits aplha and beta by immunoblotting and by real-time RT-PCR. Total myocardial CK activity was also assessed. Protein levels of CKM and CK activity were reduced in all three cardiomyopathy groups. However, total CK activity, as well as ATP synthase alpha chain protein levels, were significantly lower in CCC samples than IC and IDC samples. CCC myocardium displayed selective reduction of protein levels and activity of enzymes crucial for maintaining cytoplasmic ATP levels. Conclusions/Significance The selective impairment of the CK system may be associated to the loss of inotropic reserve observed in CCC. Reduction of ATP synthase alpha levels is consistent with a decrease in myocardial ATP generation through oxidative phosphorylation. Together, these results suggest that the energetic deficit is more intense in the myocardium of CCC patients than in the other tested dilated cardiomyopathies. Chronic Chagas disease cardiomyopathy (CCC) affects millions in endemic areas and is presenting in growing numbers in the USA and European countries due to migration currents. Clinical progression, length of survival and overall prognosis are significantly worse in CCC patients when compared to patients with dilated cardiomyopathy of non-inflammatory etiology. Impairment of energy metabolism seems to play a role in heart failure due to cardiomyopathies. Herein, we have analyzed energy metabolism enzymes in myocardium samples of CCC patients comparing to other non-inflammatory cardiomyopathies. We found that myocardial tissue from CCC patients displays a significant reduction of both myocardial protein levels of ATP synthase alpha and creatine kinase enzyme activity, in comparison to control heart samples, as well as idiopathic dilated cardiomyopathy and ischemic cardiomyopathy. Our results suggest that CCC myocardium displays a selective energetic deficit, which may play a role in the reduced heart function observed in such patients.
Collapse
Affiliation(s)
- Priscila Camillo Teixeira
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | | | - Alfredo Inácio Fiorelli
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Luiz Alberto Benvenuti
- Division of Pathology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Noedir Antonio Stolf
- Division of Surgery, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, São Paulo, Brazil
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
50
|
Fabbro DL, Olivera V, Bizai ML, Denner S, Diez C, Marcipar I, Mancipar I, Streiger M, Arias E, del Barco M, Mendicino D, Bottasso O. Humoral immune response against P2β from Trypanosoma cruzi in persons with chronic Chagas disease: its relationship with treatment against parasites and myocardial damage. Am J Trop Med Hyg 2011; 84:575-80. [PMID: 21460013 DOI: 10.4269/ajtmh.2011.10-0261] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
We investigated the relationship between potentially pathogenic antibodies against a Trypanosoma cruzi ribosomal protein (P2β) and the evolution of Chagas disease and the effect of trypanocidal treatment on these variables. Seventy-eight patients with chronic Chagas disease who were followed-up for more than 20 years were divided into three groups: 30 asymptomatic persons undergoing specific treatment (group A), 37 asymptomatic persons not undergoing specific treatment (group B), and 11 patients with chronic chagasic cardiomyopathy (CCC) who were not treated. Five patients in group B showed evolution to myocardial abnormalities. Among persons with CCC, six showed no changes; the remaining persons showed progression of cardiac involvement. Levels of antibodies to P2β in persons in group A decreased from their initial values. This finding was not observed in persons in groups B and C. Comparisons at the end of the follow-up showed lower amounts of antibodies to P2β in groups A and C. These findings support the benefits of specific treatment during chronic infection.
Collapse
Affiliation(s)
- Diana L Fabbro
- Centro de Investigaciones Sobre Endemias Nacionales, Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|