1
|
Staneviciute A, B Scariot D, Liu YG, Qian Y, Sharma S, Mbaye EHA, Almunif S, Engman DM, Scott EA. Dual Treatment of Chronic Chagasic Cardiomyopathy and Parasitic Burden via Combination Nanotherapy. ACS NANO 2025; 19:16650-16664. [PMID: 40279477 DOI: 10.1021/acsnano.5c00669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
In chronic Chagas disease, the persistence of the protozoan Trypanosoma cruzi (T. cruzi) is associated with an extensive inflammatory response that impacts cardiac function. The standard treatment, oral benznidazole, effectively targets the parasitic burden but does not address the chronic inflammation nor prevent the progression of severe cardiomyopathies. This presents an inherent immunotherapeutic challenge, as implementing an anti-inflammatory approach can have the unwanted effect of inhibiting beneficial parasite-specific immunity. Here, we investigated a combination therapy approach using benznidazole and immunomodulatory rapamycin-loaded poly(ethylene glycol)-b-poly(propylene sulfide) polymersome nanocarriers in a chronic Chagas disease murine model with cardiac abnormalities. The combined treatment demonstrated effective management of both inflammation and parasitic burden at systemic and local levels. No systemic reactivation of T. cruzi infection was observed, along with cardioprotective immunomodulatory effects through the modulation of cytokines, management of parasitic burden, and improved cardiac function based on electrocardiography assessment. The combination treatment enhanced a protective cytokine response in the heart, characterized by increased anti-inflammatory IL-10 levels, achieving greater effects than standard benznidazole treatment, and normalized TNF-α levels. Localized immunomodulatory effects, along with parasitic burden control, extended to other solid tissues relevant to parasite pathology and reservoirs. These findings highlight the therapeutic potential of modulating the immune response in chronic Chagas disease with rapamycin polymersomes and emphasize the importance of precise treatment timing in the strategy's efficacy.
Collapse
Affiliation(s)
- Austeja Staneviciute
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Debora B Scariot
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Yu-Gang Liu
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Yuan Qian
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Swagat Sharma
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - El Hadji Arona Mbaye
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Sultan Almunif
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
- Bioengineering Institute, King Abdulaziz City for Science and Technology, Riydah 12354, Saudi Arabia
| | - David M Engman
- Department of Pathology, Northwestern University, Chicago, Illinois 60611, United States
| | - Evan A Scott
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| |
Collapse
|
2
|
Senna Dos Santos G, Fonseca BDR, Sousa FSS, Seixas FK, Borsuk S. Evaluation of the immunogenic potential of recombinant Mycobacterium bovis BCG expressing the ASP-2 and TC24 proteins from Trypanosoma cruzi. Acta Trop 2025; 263:107569. [PMID: 40010681 DOI: 10.1016/j.actatropica.2025.107569] [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: 11/05/2024] [Revised: 01/28/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Although drugs have been employed over the past years to treat Chagas disease, they work mostly on the acute phase of infection, where diagnosis is hardly ever made, and present a plethora of negative side effects that ends in discontinuation of treatment. Consequently, to deal with this disease, prophylaxis seems to be a better strategy, with recombinant subunit vaccines showing promising results. Among those, Mycobaterium bovis Bacillus Calmette-Guérin (BCG) has recently been employed as vector for delivering T. cruzi antigens with positive results on immune response stimulus and protection against the infection. Following this perspective, this study aimed to characterize the immune response elicited by recombinant BCG expressing a fraction of the amastigote surface protein 2 (ASP-2) and the 24 kDa flagellar calcium-binding protein (TC24) of T. cruzi. To accomplish this, four groups of BALB/c female mice (n = 10) were vaccinated with 0.9% saline solution (Group 1), non-transformed BCG Pasteur (Group 2), rBCG/pUS977/ASP-2 (Group 3) or rBCG/pUS977/TC24 (Group 4). Cellular responses, assessed by cytokine expression from cultured and protein stimulated splenocytes, were statistically higher for both vaccinal formulations when compared with basal levels (Group 1) and non-transformed BCG (Group 2). Group 3 achieved better results for interleukins 10 and 17, while interferon γ was greatly stimulated by vaccination with Group 4. Even though further analyses are needed to evaluate the full efficacy of the constructions, the here presented results exhibit the potential of BCG vectored vaccines in eliciting Th1/Th2/Th17 mixed immune responses.
Collapse
Affiliation(s)
- Guilherme Senna Dos Santos
- Laboratório de Biotecnologia Infecto-parasitária, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-900, RS, Brasil
| | - Bárbara da Rocha Fonseca
- Laboratório de Biotecnologia Infecto-parasitária, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-900, RS, Brasil
| | - Fernanda Severo Sabedra Sousa
- Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-900, RS, Brasil
| | - Fabiana Kommling Seixas
- Laboratório de Biotecnologia do Câncer, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-900, RS, Brasil
| | - Sibele Borsuk
- Laboratório de Biotecnologia Infecto-parasitária, Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas 96010-900, RS, Brasil.
| |
Collapse
|
3
|
Bocchi EA, Veiga Guimarães G, Espinoza Romero C, Sato PK, de Freitas VLT, Yamashiro Kanashiro EH, Furuchó CR, Das Dores Cruz F, Shimoda Nakanishi É, Busser FD, Bezerra RC, Westphalen EVN, Cisotto Rocha M, Shikanai Yasuda MA. Assessment of biomarkers and clinical parameters as predictors of survival in patients with chagasic heart failure. PLoS Negl Trop Dis 2023; 17:e0011847. [PMID: 38109427 PMCID: PMC10760923 DOI: 10.1371/journal.pntd.0011847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/02/2024] [Accepted: 12/06/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Chagas disease, endemic in Latin America and spreading globally due to emigration, has a significant health burden, particularly in relation to chagasic heart failure (HF). Chagasic cardiomyopathy (CCM) is characterized by chronic inflammatory myocardial disease. This study aimed to identify inflammatory parameters and biomarkers that could aid in the management of patients with chagasic HF. METHODS AND FINDINGS A cohort study was conducted at a tertiary cardiology single-center over a mean follow-up period of 2.4 years. The study included patients with HF secondary to CCM enrolled between October 2013 and July 2017. Various clinical parameters, echocardiography findings, parasitemia status, brain natriuretic peptide (BNP) and troponin T (TnT) levels, and inflammatory biomarkers (IL-6, IL-10, IL-12p70, IL-17A, adiponectin, and IFN-γ) were assessed. The study encompassed a cohort of 103 patients, with a median age of 53 years and 70% being male. The left ventricular ejection fraction (LVEF) was 28%, with 40% of patients classified as NYHA II functional class. The median BNP level was 291 pg/ml. The observed mortality rate during the study period was 38.8%. Predictors of lower survival were identified as elevated levels of BNP, TnT, reduced LVEF, and increased adiponectin (thresholds: BNP > 309 pg/ml, TnT > 27.5 ng/ml, LVEF < 25.5%, adiponectin > 38 μg/mL). Notably, there was no evidence indicating a relationship between parasitemia and the inflammatory parameters with lower survival in these patients, including INF-γ, IL-6, IL-10, IL12-(p70), and IL17a. CONCLUSION Despite the presence of a chronic inflammatory process, the evaluated inflammatory biomarkers in this cohort were not predictive of survival in patients with chagasic HF with reduced ejection fraction (HFrEF). However, reduced LVEF, elevated BNP, adiponectin levels, and troponin T were identified as predictors of lower survival in these patients.
Collapse
Affiliation(s)
- Edimar Alcides Bocchi
- Heart Failure Clinics, Instituto do Coração Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Guilherme Veiga Guimarães
- Heart Failure Clinics, Instituto do Coração Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Cristhian Espinoza Romero
- Heart Failure Clinics, Instituto do Coração Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Paula Keiko Sato
- Laboratory of Medical Investigation in Immunology (LIM-48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
| | - Vera Lúcia Teixeira de Freitas
- Laboratory of Medical Investigation in Immunology (LIM-48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
- Departament of Infectious Diseases, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| | - Edite Hatsumi Yamashiro Kanashiro
- Laboratory of Medical Investigation in Immunology (LIM-48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
- Departament of Infectious Diseases, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| | - Célia Regina Furuchó
- Laboratory of Medical Investigation in Immunology (LIM-48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
| | - Fatima Das Dores Cruz
- Heart Failure Clinics, Instituto do Coração Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | | | - Felipe Delatorre Busser
- Departament of Infectious Diseases, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| | - Rita Cristina Bezerra
- Laboratory of Medical Investigation in Parasitology (LIM-46), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
| | | | - Mussya Cisotto Rocha
- Laboratory of Medical Investigation in Immunology (LIM-48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
| | - Maria Aparecida Shikanai Yasuda
- Laboratory of Medical Investigation in Immunology (LIM-48), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, Brazil
- Departament of Infectious Diseases, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Liu Z, Ulrich vonBargen R, Kendricks AL, Wheeler K, Leão AC, Sankaranarayanan K, Dean DA, Kane SS, Hossain E, Pollet J, Bottazzi ME, Hotez PJ, Jones KM, McCall LI. Localized cardiac small molecule trajectories and persistent chemical sequelae in experimental Chagas disease. Nat Commun 2023; 14:6769. [PMID: 37880260 PMCID: PMC10600178 DOI: 10.1038/s41467-023-42247-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 10/04/2023] [Indexed: 10/27/2023] Open
Abstract
Post-infectious conditions present major health burdens but remain poorly understood. In Chagas disease (CD), caused by Trypanosoma cruzi parasites, antiparasitic agents that successfully clear T. cruzi do not always improve clinical outcomes. In this study, we reveal differential small molecule trajectories between cardiac regions during chronic T. cruzi infection, matching with characteristic CD apical aneurysm sites. Incomplete, region-specific, cardiac small molecule restoration is observed in animals treated with the antiparasitic benznidazole. In contrast, superior restoration of the cardiac small molecule profile is observed for a combination treatment of reduced-dose benznidazole plus an immunotherapy, even with less parasite burden reduction. Overall, these results reveal molecular mechanisms of CD treatment based on simultaneous effects on the pathogen and on host small molecule responses, and expand our understanding of clinical treatment failure in CD. This link between infection and subsequent persistent small molecule perturbation broadens our understanding of infectious disease sequelae.
Collapse
Affiliation(s)
- Zongyuan Liu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Rebecca Ulrich vonBargen
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
- Department of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | | | - Kate Wheeler
- Department of Biology, University of Oklahoma, Norman, OK, USA
| | - Ana Carolina Leão
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Krithivasan Sankaranarayanan
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA
| | - Danya A Dean
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Shelley S Kane
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Ekram Hossain
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA
| | - Jeroen Pollet
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Maria Elena Bottazzi
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Hotez
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kathryn M Jones
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA.
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, OK, USA.
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, OK, USA.
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA, USA.
| |
Collapse
|
5
|
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] [Received: 06/27/2023] [Accepted: 09/19/2023] [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
|
6
|
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: 25] [Impact Index Per Article: 12.5] [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
|
7
|
Cutshaw MK, Sciaudone M, Bowman NM. Risk Factors for Progression to Chronic Chagas Cardiomyopathy: A Systematic Review and Meta-Analysis. Am J Trop Med Hyg 2023; 108:791-800. [PMID: 36848894 PMCID: PMC10076993 DOI: 10.4269/ajtmh.22-0630] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/19/2022] [Indexed: 03/01/2023] Open
Abstract
Approximately one-third of people with chronic Trypanosoma cruzi infection develop Chagas cardiomyopathy, which carries a poor prognosis. Accurate prediction of which individuals will go on to develop Chagas cardiomyopathy remains elusive. We performed a systematic review of literature comparing characteristics of individuals with chronic Chagas disease with or without evidence of cardiomyopathy. Studies were not excluded on the basis of language or publication date. Our review yielded a total of 311 relevant publications. We further examined the subset of 170 studies with data regarding individual age, sex, or parasite load. A meta-analysis of 106 eligible studies indicated that male sex was associated with having Chagas cardiomyopathy (Hedge's g: 1.56, 95% CI: 1.07-2.04), and a meta-analysis of 91 eligible studies indicated that older age was associated with having Chagas cardiomyopathy (Hedge's g: 0.66, 95% CI: 0.41-0.91). A meta-analysis of four eligible studies did not find an association between parasite load and disease state. This study provides the first systematic review to assess whether age, sex, and parasite load are associated with Chagas cardiomyopathy. Our findings suggest that older and male patients with Chagas disease are more likely to have cardiomyopathy, although we are unable to identify causal relationships due to the high heterogeneity and predominantly retrospective study designs in the current literature. Prospective, multidecade studies are needed to better characterize the clinical course of Chagas disease and identify risk factors for progression to Chagas cardiomyopathy.
Collapse
Affiliation(s)
| | - Michael Sciaudone
- Section of Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana
| | - Natalie M. Bowman
- Division of Infectious Diseases, Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
8
|
Liu Z, Ulrich R, Kendricks AL, Wheeler K, Leão AC, Pollet J, Bottazzi ME, Hotez P, Gusovsky F, Jones KM, McCall LI. Localized cardiac metabolic trajectories and post-infectious metabolic sequelae in experimental Chagas disease. RESEARCH SQUARE 2023:rs.3.rs-2497474. [PMID: 36711878 PMCID: PMC9882638 DOI: 10.21203/rs.3.rs-2497474/v1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Post-infectious conditions, where clinical symptoms fail to resolve even after pathogen clearance, present major health burdens. However, the mechanisms involved remain poorly understood. In Chagas disease (CD), caused by the parasite Trypanosoma cruzi, antiparasitic agents can clear T. cruzi but late-stage treatment does not improve clinical cardiac outcomes. In this study, we revealed differential metabolic trajectories of cardiac regions during T. cruzi infection, matching sites of clinical symptoms. Incomplete, region-specific, cardiac metabolic restoration was observed in animals treated with the antiparasitic benznidazole, even though parasites were successfully cleared. In contrast, superior metabolic restoration was observed for a combination treatment of reduced-dose benznidazole plus an immunotherapy (Tc24-C4 T. cruzi flagellar protein and TLR4 agonist adjuvant), even though parasite burden reduction was lower. Overall, these results provide a mechanism to explain prior clinical treatment failures in CD and to test novel candidate treatment regimens. More broadly, our results demonstrate a link between persistent metabolic perturbation and post-infectious conditions, with broad implications for our understanding of post-infectious disease sequelae.
Collapse
Affiliation(s)
- Zongyuan Liu
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Rebecca Ulrich
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, United States of America
| | - April L. Kendricks
- Southern Star Medical Research Institute, Houston, TX, United States of America
| | - Kate Wheeler
- Department of Biology, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Ana Carolina Leão
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States of America
| | - Jeroen Pollet
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States of America
| | - Maria Elena Bottazzi
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Peter Hotez
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | | | - Kathryn M. Jones
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States of America
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma, United States of America
| |
Collapse
|
9
|
Campos-Estrada C, Urarte E, Denegri M, Villalón L, González-Herrera F, Kemmerling U, Maya JD. Effect of statins on inflammation and cardiac function in patients with chronic Chagas disease: A protocol for pathophysiological studies in a multicenter, placebo-controlled, proof-of-concept phase II trial. PLoS One 2023; 18:e0280335. [PMID: 36638112 PMCID: PMC9838836 DOI: 10.1371/journal.pone.0280335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/23/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Cardiac complications, including heart failure and arrhythmias, are the leading causes of disability and death in Chagas disease (CD). CD, caused by the Trypanosoma cruzi parasite, afflicts 7 million people in Latin America, and its incidence is increasing in non-endemic countries due to migration. The cardiac involvement is explained by parasite-dependent, immune-mediated myocardial injury, microvascular abnormalities, and ischemia. Current treatment of early CD includes the administration of nifurtimox and benznidazole. However, their efficacy is low in the chronic phase and may induce severe adverse events, forcing therapy to halt. Therefore, finding innovative approaches to treat this life-threatening tropical disease is of utmost importance. Thus, improving the efficacy of the current antichagasic drugs by modifying the inflammatory response would render the current treatment more effective. It has been reported that, in mice, simvastatin decreases cardiac inflammation and endothelial activation, and improves cardiac function, effects that require clinical confirmation. OBJECTIVE The study aims to analyze whether two doses of Atorvastatin, administered after CD treatment is completed, are safe and more efficacious than the antiparasitic drugs alone in reducing general inflammation and improving endothelial and cardiac functions in a proof-of-concept, placebo-controlled phase II trial. METHODS 300 subjects will be recruited from four Chilean hospitals with an active Program for the Control of Chagas Disease. 40 or 80 mg/day of atorvastatin or placebo will be administered after completion of the antichagasic therapy. The patients will be followed up for 12 months. Efficacy will be determined by measuring changes in plasma levels of anti-inflammatory and pro-inflammatory cytokines, soluble cell adhesion molecules, BNP, and cTnT. Also, the resting 12-lead ECG and a 2D-echocardiogram will be obtained to evaluate cardiac function. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04984616.
Collapse
Affiliation(s)
| | - Edurne Urarte
- Departamento de Pediatría y Cirugía Infantil Occidente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marisol Denegri
- Departamento de Pediatría y Cirugía Infantil Occidente, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Litzi Villalón
- Servicio de Salud Viña del Mar-Quillota, Hospital Gustavo Fricke, Viña del Mar, Chile
| | - Fabiola González-Herrera
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Programa de Biología Integrativa, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail: (JDM); (UK)
| | - Juan D. Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail: (JDM); (UK)
| |
Collapse
|
10
|
Paula AT, Ribeiro KVG, Cardoso KF, Bastos DSS, Santos EC, Novaes RD, Cardoso SA, Oliveira LL. Protective immunity triggered by ectonucleoside triphosphate diphosphohydrolase-based biopharmaceuticals attenuates cardiac parasitism and prevents mortality in Trypanosoma cruzi infection. Bioorg Med Chem 2022; 72:116966. [PMID: 35998390 DOI: 10.1016/j.bmc.2022.116966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/04/2022] [Accepted: 08/12/2022] [Indexed: 11/26/2022]
Abstract
Chagas disease is a potentially fatal infection in 21 endemic Latin America countries for which the effectiveness of reference antiparasitic chemotherapy is limited. Thus, we developed three biopharmaceuticals and evaluated the effectiveness of different immunization strategies (recombinant protein NTPDase-1 [rNTPDase-1], DNA plasmid encoding Trypanosoma cruzi NTPDase-1 [TcNTPDase-1] and DNA-NTPDase-1 prime/rNTPDase-1 boost [Prime-boost]) based on the surface ecto-nucleoside triphosphate diphosphohydrolase (ecto-NTPDase) enzyme of T. cruzi in animals challenged with a virulent strain (Y) of this parasite. BALB/c mice were immunized three times at 30 days intervals, challenged with T. cruzi 15 days after the last immunization, and euthanized 30 days after T. cruzi challenge. Our results showed limited polarization of specific anti-ecto-NTPDase immunoglobulins in mice receiving both immunization protocols. Conversely, the Prime-boost strategy stimulated the Th1 protective phenotype, upregulating TNF-α and downregulating IL-10 production while increasing the activation/distribution of CD3+/CD8+, CD4+/CD44hi and CD8+/CD44hi/CD62L cells in immunized and infected mice. Furthermore, IL-6 and IL10 levels were reduced, while the distribution of CD4+/CD44hi and CD3+/CD8+ cells was increased from rNTPDase-1 and DNA-NTPDase1-based immunization strategies. Animals receiving DNA-NTPDase1 and Prime-boost protocols before T. cruzi challenged exhibited an enhanced immunological response associated with IL-17 upregulation and remarkable downregulation of heart parasitism (T. cruzi DNA) and mortality. These findings indicated that NTPDase-1 with Prime-boost strategy induced a protective and sustained Th17 response, enhancing host resistance against T. cruzi. Thus, ecto-NTPDase is a potentially relevant and applicable in the development of biopharmaceuticals with greater immunoprophylactic potential for Chagas disease.
Collapse
Affiliation(s)
| | | | | | | | - Eliziária Cardoso Santos
- School of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, MG, Brazil
| | - Rômulo Dias Novaes
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-001, MG, Brazil
| | - Silvia Almeida Cardoso
- Department of Medicine and Nursing, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
| | | |
Collapse
|
11
|
Biomakers in Chronic Chagas Cardiomyopathy. Microorganisms 2022; 10:microorganisms10081602. [PMID: 36014020 PMCID: PMC9412448 DOI: 10.3390/microorganisms10081602] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/22/2022] [Accepted: 07/29/2022] [Indexed: 11/17/2022] Open
Abstract
The primary objective was to observe the relationship between serum levels of BNP, Ca-125, C-reactive protein and uric acid as prognostic and functional markers in patients with chronic Chagas cardiomyopathy (CCC). Circulating levels of cytokines: IL-1β, TNFα, IL-10, IL6, IL-8 and IL-12 were determined and investigated regarding their association with hemodynamic parameters, clinical signs of heart failure and outcome. Chagas is still a neglected disease that affects numerous individuals, many of them in their most productive years. CCC with left ventricular dysfunction is the most severe presentation of Chagas Disease. BNP is a well-recognized prognostic and clinical biomarker, not only in chronic heart failure patients but also in patients with CCC. Previous studies have shown Ca-125, C-reactive protein, and uric acid to be potentially good prognostic markers in heart failure (HF). Fifty patients with left ventricular fraction less (LVEF) than 55% were selected and followed for a mean period of 18 ± 8.3 months. Patient’s mean age was 43.42 ± 10.3 years (32 male), their BNP was 293 (160–530) pg/mL, Ca-125 8.5 (5.5–16.75) U/mL, uric acid 6.2 ± 2 mg/dL, and C- reactive protein 4.5 (4.5–7.3) mg/L. Patients who had LVEF less than 35% had higher BNP (p = 0.0023), Ca-125 (p = 0.027) and uric acid (p = 0.01) serum levels. Patients who died also showed higher BNP (p = 0.01), uric acid (p = 0.05) and a trend towards higher Ca-125 serum levels (p = 0.056). All markers: BNP, Ca-125, uric acid and C-reactive had good predictability of death in Cox-regression univariate analysis, however, not on the final multivariate model. Of the inflammatory cytokines, IL-8 and IL-12 showed a relation to LVEF of less than 35%. IL-12 was related to adverse cardiovascular events and non-survival. IL-1β was a good predictor of mortality in the final Cox regression model. Determination of Ca-125, uric acid levels and C-reactive protein may add useful clinical and prognostic information and may help clinical decision making for patients with CCC.
Collapse
|
12
|
Oliveira TGM, Venturini G, Alvim JM, Feijó LL, Dinardo CL, Sabino EC, Seidman JG, Seidman CE, Krieger JE, Pereira AC. Different Transcriptomic Response to T. cruzi Infection in hiPSC-Derived Cardiomyocytes From Chagas Disease Patients With and Without Chronic Cardiomyopathy. Front Cell Infect Microbiol 2022; 12:904747. [PMID: 35873155 PMCID: PMC9301326 DOI: 10.3389/fcimb.2022.904747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas disease is a tropical zoonosis caused by Trypanosoma cruzi. After infection, the host present an acute phase, usually asymptomatic, in which an extensive parasite proliferation and intense innate immune activity occurs, followed by a chronic phase, characterized by low parasitemia and development of specific immunity. Most individuals in the chronic phase remain without symptoms or organ damage, a state called indeterminate IND form. However, 20 to 40% of individuals develop cardiac or gastrointestinal complications at any time in life. Cardiomyocytes have an important role in the development of Chronic Chagas Cardiomyopathy (CCC) due to transcriptional and metabolic alterations that are crucial for the parasite survival and replication. However, it still not clear why some infected individuals progress to a cardiomyopathy phase, while others remain asymptomatic. In this work, we used hiPSCs-derived cardiomyocytes (hiPSC-CM) to investigate patterns of infection, proliferation and transcriptional response in IND and CCC patients. Our data show that T. cruzi infection and proliferation efficiency do not differ significantly in PBMCs and hiPSC-CM from both groups. However, RNA-seq analysis in hiPSC-CM infected for 24 hours showed a significantly different transcriptional response to the parasite in cells from IND or CCC patients. Cardiomyocytes from IND showed significant differences in the expression of genes related to antigen processing and presentation, as well as, immune co-stimulatory molecules. Furthermore, the downregulation of collagen production genes and extracellular matrix components was significantly different in these cells. Cardiomyocytes from CCC, in turn, showed increased expression of mTORC1 pathway and unfolded protein response genes, both associated to increased intracellular ROS production. These data point to a differential pattern of response, determined by baseline genetic differences between groups, which may have an impact on the development of a chronic outcome with or without the presentation of cardiac symptoms.
Collapse
Affiliation(s)
- Theo G. M. Oliveira
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Fundação Pró-Sangue Hemocentro de São Paulo, Divisão de Pesquisa – São Paulo, SP, Brazil
- *Correspondence: Theo G. M. Oliveira,
| | - Gabriela Venturini
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Genetics Department, Harvard Medical School, MA, United States
| | - Juliana M. Alvim
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
| | - Larissa L. Feijó
- Fundação Pró-Sangue Hemocentro de São Paulo, Divisão de Pesquisa – São Paulo, SP, Brazil
| | - Carla L. Dinardo
- Fundação Pró-Sangue Hemocentro de São Paulo, Divisão de Pesquisa – São Paulo, SP, Brazil
| | - Ester C. Sabino
- Instituto do Medicina Tropical (IMT), Universidade de São Paulo – São Paulo, SP, Brazil
| | | | - Christine E. Seidman
- Genetics Department, Harvard Medical School, MA, United States
- Cardiovascular Division, Brigham and Women’s Hospital, & Harvard Medical School, Boston, MA, United States
- Howard Hughes Medical Institute, Chevy Chase, MD, United States
| | - Jose E. Krieger
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
| | - Alexandre C. Pereira
- Laboratório de Genética e Cardiologia Molecular, Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Instituto do Coração (InCor), Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HC/FMUSP), São Paulo, Brazil
- Genetics Department, Harvard Medical School, MA, United States
| |
Collapse
|
13
|
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: 6] [Impact Index Per Article: 2.0] [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
|
14
|
Pinto BF, Medeiros NI, Teixeira-Carvalho A, Fiuza JA, Eloi-Santos SM, Nunes MCP, Silva SA, Fontes-Cal TCM, Belchior-Bezerra M, Dutra WO, Correa-Oliveira R, Gomes JAS. Modulation of Regulatory T Cells Activity by Distinct CD80 and CD86 Interactions With CD28/CTLA-4 in Chagas Cardiomyopathy. Front Cardiovasc Med 2022; 9:750876. [PMID: 35665256 PMCID: PMC9162138 DOI: 10.3389/fcvm.2022.750876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 04/08/2022] [Indexed: 11/16/2022] Open
Abstract
Chagas cardiomyopathy is the symptomatic cardiac clinical form (CARD) of the chronic phase of Chagas disease caused by Trypanosoma cruzi infection. It was described as the most fibrosing cardiomyopathies, affecting approximately 30% of patients during the chronic phase. Other less frequent symptomatic clinical forms have also been described. However, most patients who progress to the chronic form develop the indeterminate clinical form (IND), may remain asymptomatic for life, or develop some cardiac damage. Some mechanisms involved in the etiology of the clinical forms of Chagas disease have been investigated. To characterize the contribution of CD80 and CD86 co-stimulatory molecules in the activation of different CD4+ (Th1, Th2, Th17, and Treg) and CD8+ T lymphocyte subsets, we used blocking antibodies for CD80 and CD86 receptors of peripheral blood mononuclear cells (PBMC) in cultures with T. cruzi antigens from non-infected (NI), IND, and CARD individuals. We demonstrated a higher frequency of CD8+ CD25+ T lymphocytes and CD8+ Treg cells after anti-CD80 antibody blockade only in the CARD group. In contrast, a lower frequency of CD4+ Treg lymphocytes after anti-CD86 antibody blockade was found only in IND patients. A higher frequency of CD4+ Treg CD28+ lymphocytes, as well as an association between CD4+ Treg lymphocytes and CD28+ expression on CD4+ Treg cells in the CARD group, but not in IND patients, and once again only after anti-CD80 antibody blockade, was observed. We proposed that Treg cells from IND patients could be activated via CD86-CTLA-4 interaction, leading to modulation of the immune response only in asymptomatic patients with Chagas disease, while CD80 may be involved in the proliferation control of T CD8+ lymphocytes, as also in the modulation of regulatory cell activation via CD28 receptor. For the first time, our data highlight the role of CD80 in modulation of Treg lymphocytes activation in patients with CARD, highlighting a key molecule in the development of Chagas cardiomyopathy.
Collapse
Affiliation(s)
- Bruna F. Pinto
- Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nayara I. Medeiros
- Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto René Rachou, Fundação Oswaldo Cruz–FIOCRUZ, Belo Horizonte, Brazil
| | | | - Jacqueline A. Fiuza
- Instituto René Rachou, Fundação Oswaldo Cruz–FIOCRUZ, Belo Horizonte, Brazil
| | | | - Maria C. P. Nunes
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Silvana A. Silva
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Tereza C. M. Fontes-Cal
- Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mayara Belchior-Bezerra
- Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Walderez O. Dutra
- Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia Doenças Tropicais, Belo Horizonte, Brazil
| | - Rodrigo Correa-Oliveira
- Instituto René Rachou, Fundação Oswaldo Cruz–FIOCRUZ, Belo Horizonte, Brazil
- Instituto Nacional de Ciência e Tecnologia Doenças Tropicais, Belo Horizonte, Brazil
| | - Juliana A. S. Gomes
- Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Juliana A. S. Gomes,
| |
Collapse
|
15
|
Gómez I, Thomas MC, Palacios G, Egui A, Carrilero B, Simón M, Valladares B, Segovia M, Carmelo E, López MC. Differential Expression of Immune Response Genes in Asymptomatic Chronic Chagas Disease Patients Versus Healthy Subjects. Front Cell Infect Microbiol 2021; 11:722984. [PMID: 34552885 PMCID: PMC8450343 DOI: 10.3389/fcimb.2021.722984] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/12/2021] [Indexed: 11/30/2022] Open
Abstract
Infection by the Trypanosoma cruzi parasite causes Chagas disease and triggers multiple immune mechanisms in the host to combat the pathogen. Chagas disease has a variable clinical presentation and progression, producing in the chronic phase a fragile balance between the host immune response and parasite replication that keeps patients in a clinically silent asymptomatic stage for years. Since the parasite is intracellular and replicates within cells, the cell-mediated response of the host adaptive immunity plays a critical role. This function is mainly orchestrated by T lymphocytes, which recognize parasite antigens and promote specific functions to control the infection. However, little is known about the immunological markers associated with this asymptomatic stage of the disease. In this large-scale analysis, the differential expression of 106 immune system-related genes has been analyzed using high-throughput qPCR in T. cruzi antigen-stimulated PBMC from chronic Chagas disease patients with indeterminate form (IND) and healthy donors (HD) from endemic and non-endemic areas of Chagas disease. This analysis revealed that there were no differences in the expression level of most genes under study between healthy donors from endemic and non-endemic areas determined by PCA and differential gene expression analysis. Instead, PCA revealed the existence of different expression profiles between IND patients and HD (p < 0.0001), dependent on the 32 genes included in PC1. Differential gene expression analysis also revealed 23 upregulated genes (expression fold change > 2) and 11 downregulated genes (expression fold change < 0.5) in IND patients versus HD. Enrichment analysis showed that several upregulated genes in IND patients participate in relevant immunological pathways such as antigen-dependent B cell activation, stress induction of HSP regulation, NO2-dependent IL12 pathway in NK cells, and cytokine-inflammatory response. The antigen-specific differential gene expression profile detected in these patients and the relevant immunological pathways that seem to be activated could represent potential biomarkers of the asymptomatic form of Chagas disease, helpful to diagnosis and infection control.
Collapse
Affiliation(s)
- Inmaculada Gómez
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - M Carmen Thomas
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Génesis Palacios
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| | - Adriana Egui
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Bartolomé Carrilero
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Marina Simón
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Basilio Valladares
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain.,Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, La Laguna, Spain
| | - Manuel Segovia
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Emma Carmelo
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain.,Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, La Laguna, Spain
| | - Manuel Carlos López
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
16
|
Choudhuri S, Rios L, Vázquez-Chagoyán JC, Garg NJ. Oxidative stress implications for therapeutic vaccine development against Chagas disease. Expert Rev Vaccines 2021; 20:1395-1406. [PMID: 34406892 DOI: 10.1080/14760584.2021.1969230] [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: 10/20/2022]
Abstract
INTRODUCTION Pathogenesis of Chagas disease (CD) caused by the protozoan parasite Trypanosoma cruzi (T. cruzi) involves chronic oxidative and inflammatory stress. In this review, we discuss the research efforts in therapeutic vaccine development to date and the potential challenges imposed by oxidative stress in achieving an efficient therapeutic vaccine against CD. AREAS COVERED This review covers the immune and nonimmune mechanisms of reactive oxygen species production and immune response patterns during T. cruzi infection in CD. A discussion on immunotherapy development efforts, the efficacy of antigen-based immune therapies against T. cruzi, and the role of antioxidants as adjuvants is discussed to provide promising insights to developing future treatment strategies against CD. EXPERT OPINION Administration of therapeutic vaccines can be a good option to confront persistent parasitemia in CD by achieving a rapid, short-lived stimulation of type 1 cell-mediated immunity. At the same time, adjunct therapies could play a critical role in the preservation of mitochondrial metabolism and cardiac muscle contractility in CD. We propose combined therapy with antigen-based vaccine and small molecules to control the pathological oxidative insult would be effective in the conservation of cardiac structure and function in CD.
Collapse
Affiliation(s)
- Subhadip Choudhuri
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Lizette Rios
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Juan Carlos Vázquez-Chagoyán
- Centro de Investigación y Estudios Avanzados En Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca, México
| | - Nisha Jain Garg
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Tx, USA
| |
Collapse
|
17
|
Girard MC, Ossowski MS, Muñoz-Calderón A, Fernández M, Hernández-Vásquez Y, Chadi R, Gómez KA. Trypanosoma cruzi Induces Regulatory B Cell Alterations in Patients With Chronic Chagas Disease. Front Cell Infect Microbiol 2021; 11:723549. [PMID: 34458163 PMCID: PMC8387560 DOI: 10.3389/fcimb.2021.723549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/21/2021] [Indexed: 12/02/2022] Open
Abstract
The clinical evolution of patients with chronic Chagas disease (CCD) is mainly associated with an excessive inflammation and a defective immunomodulatory profile caused by the interaction between T. cruzi and the host. Regulatory B (Breg) cells exert immune suppression mostly through IL-10 production (B10 cells), but also through IL-10-independent mechanisms. Previously, we demonstrated that CCD patients with cardiomyopathy show changes in the ex vivo Breg cell phenotypic distribution although maintain IL-10 production capacity. Here, we sought to identify potential alterations on Breg cells upon in vitro stimulation. Isolated B cells from CCD patients with or without cardiomyopathy and non-infected (NI) donors were stimulated with T. cruzi lysate or CpG + CD40L, and characterized by flow cytometry based on the expression of CD24, CD27, CD38, and the regulatory molecules IL-10 and PD-L1. IL-10 and IL-17 secretion in the supernatant of B cells was evaluated by ELISA. Data showed that T. cruzi stimulation diminished the expression of CD24 and CD38 on CD27- B cells while reducing the percentage of CD24high inside CD27+ B cells. Furthermore, T. cruzi induced a regulatory B cell phenotype by increasing B10 cells and IL-10 secretion in all the groups. The innate-like B10 cells expansion observed in patients with cardiomyopathy would be associated with CD27- B10 cell subsets, while no predominant phenotype was found in the other groups. Patients with cardiomyopathy also displayed higher IL-17 secretion levels in T. cruzi-activated B cells. CpG + CD40L stimulation revealed that B cells from CCD patients and NI donors had the same ability to differentiate into B10 cells and secrete IL-10 in vitro. Additionally, CCD patients showed an increased frequency of CD24-CD27- B cells and a reduction in the percentage of CD24highCD27+ Breg cells, which appeared to be inversely correlated with the presence of T. cruzi DNA in blood. Finally, CCD patients exhibited a higher frequency of PD-L1+ B cells in T. cruzi-stimulated samples, suggesting that IL-10-independent mechanisms could also be tangled in the control of inflammation. Altogether, our results provide evidence about the potential role of Breg cells in the immune response developed against T. cruzi and its contribution to chronic Chagas cardiomyopathy.
Collapse
Affiliation(s)
- Magalí C. Girard
- Laboratorio de Inmunología de las Infecciones por Tripanosomátidos (LIIT), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Micaela S. Ossowski
- Laboratorio de Inmunología de las Infecciones por Tripanosomátidos (LIIT), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Arturo Muñoz-Calderón
- Laboratorio de Biología Molecular de la Enfermedad de Chagas (LabMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Marisa Fernández
- Instituto Nacional de Parasitología “Dr. Mario Fatala Chabén”, Buenos Aires, Argentina
| | | | - Raúl Chadi
- Hospital General de Agudos “Dr. Ignacio Pirovano”, Buenos Aires, Argentina
| | - Karina A. Gómez
- Laboratorio de Inmunología de las Infecciones por Tripanosomátidos (LIIT), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular “Dr. Héctor N. Torres” (INGEBI), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
18
|
Hypothyroidism impairs the host immune response during the acute phase of Chagas disease. Immunobiology 2020; 225:152024. [PMID: 33227693 DOI: 10.1016/j.imbio.2020.152024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/20/2020] [Accepted: 10/18/2020] [Indexed: 11/24/2022]
Abstract
Diseases associated with thyroid hypofunction have been the subject of studies in infectious models, since several authors have demonstrated a pivotal role of iodinated hormones (thyroxine and triiodothyronine) in the modulation of immune effector responses. Using a model of hypothyroidism induced by anti-thyroid drug, we investigated the influence of hypothyroidism in the course of acute Trypanosoma cruzi infection. For this, male Hannover Wistar rats were challenged with methimazole for 21 days (0.02% in drinking water), and water for control counterparts. After confirmation of the hypothyroidism, rats were intraperitoneally challenged with 1x105 blood trypomastigotes of the Y strain of T. cruzi. Our findings suggest that hypothyroidism impairs animal weight gain, but does not affect the health of essential organs. Interestingly, infected hypothyroid animals had a significant increase in thymic cell death, with consequent drop in lymphocyte frequency in whole blood (evaluated on the 11th day of infection). Analyzing the percentage of immune cells in the spleen, we found a strong influence of hypothyroidism as a negative regulator of B cells, and antigenic ability of macrophages (RT1b expression) in the course of the experimental chagasic infection. Enhanced serum IL-17A concentration was induced by T. cruzi infection, but hypothyroidism impaired the production of this mediator as seen in infected hypothyroid animals. Taken together, our work suggests for the first time that hypothyroidism may adversely interfere with the modulation of effective immunity in the early phase of Chagas' disease.
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: 1.0] [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
|
Rada J, Donato M, Penas FN, Alba Soto C, Cevey ÁC, Pieralisi AV, Gelpi R, Mirkin GA, Goren NB. IL-10-Dependent and -Independent Mechanisms Are Involved in the Cardiac Pathology Modulation Mediated by Fenofibrate in an Experimental Model of Chagas Heart Disease. Front Immunol 2020; 11:572178. [PMID: 33072115 PMCID: PMC7541836 DOI: 10.3389/fimmu.2020.572178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
IL-10 is an anti-inflammatory cytokine that plays a significant role in the modulation of the immune response in many pathological conditions, including infectious diseases. Infection with Trypanosoma cruzi (T. cruzi), the etiological agent of Chagas disease, results in an ongoing inflammatory response that may cause heart dysfunction, ultimately leading to heart failure. Given its infectious and inflammatory nature, in this work we analyzed whether the lack of IL-10 hinders the anti-inflammatory effects of fenofibrate, a PPARα ligand, in a murine model of Chagas heart disease (CHD) using IL-10 knockout (IL-10 KO) mice. Our results show fenofibrate was able to restore the abnormal cardiac function displayed by T. cruzi-infected mice lacking IL-10. Treatment with fenofibrate reduced creatine kinase (CK) levels in sera of IL-10 KO mice infected with T. cruzi. Moreover, although fenofibrate could not modulate the inflammatory infiltrates developing in the heart, it was able to reduce the increased collagen deposition in infected IL-10 KO mice. Regarding pro-inflammatory mediators, the most significant finding was the increase in serum IL-17. These were reduced in IL-10 KO mice upon fenofibrate treatment. In agreement with this, the expression of RORγt was reduced. Infection of IL-10 KO mice increased the expression of YmI, FIZZ and Mannose Receptor (tissue healing markers) that remained unchanged upon treatment with fenofibrate. In conclusion, our work emphasizes the role of anti-inflammatory mechanisms to ameliorate heart function in CHD and shows, for the first time, that fenofibrate attains this through IL-10-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Jimena Rada
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Martín Donato
- Departamento de Patología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Fisiopatología Cardiovascular, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Federico N Penas
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Catalina Alba Soto
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones en Microbiología y Parasitología Médica, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ágata C Cevey
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Azul V Pieralisi
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ricardo Gelpi
- Departamento de Patología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Fisiopatología Cardiovascular, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gerardo A Mirkin
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones en Microbiología y Parasitología Médica, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora B Goren
- Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
21
|
Watanabe Costa R, Batista MF, Meneghelli I, Vidal RO, Nájera CA, Mendes AC, Andrade-Lima IA, da Silveira JF, Lopes LR, Ferreira LRP, Antoneli F, Bahia D. Comparative Analysis of the Secretome and Interactome of Trypanosoma cruzi and Trypanosoma rangeli Reveals Species Specific Immune Response Modulating Proteins. Front Immunol 2020; 11:1774. [PMID: 32973747 PMCID: PMC7481403 DOI: 10.3389/fimmu.2020.01774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 07/02/2020] [Indexed: 12/04/2022] Open
Abstract
Chagas disease, a zoonosis caused by the flagellate protozoan Trypanosoma cruzi, is a chronic and systemic parasitic infection that affects ~5–7 million people worldwide, mainly in Latin America. Chagas disease is an emerging public health problem due to the lack of vaccines and effective treatments. According to recent studies, several T. cruzi secreted proteins interact with the human host during cell invasion. Moreover, some comparative studies with T. rangeli, which is non-pathogenic in humans, have been performed to identify proteins directly involved in the pathogenesis of the disease. In this study, we present an integrated analysis of canonical putative secreted proteins (PSPs) from both species. Additionally, we propose an interactome with human host and gene family clusters, and a phylogenetic inference of a selected protein. In total, we identified 322 exclusively PSPs in T. cruzi and 202 in T. rangeli. Among the PSPs identified in T. cruzi, we found several trans-sialidases, mucins, MASPs, proteins with phospholipase 2 domains (PLA2-like), and proteins with Hsp70 domains (Hsp70-like) which have been previously characterized and demonstrated to be related to T. cruzi virulence. PSPs found in T. rangeli were related to protozoan metabolism, specifically carboxylases and phosphatases. Furthermore, we also identified PSPs that may interact with the human immune system, including heat shock and MASP proteins, but in a lower number compared to T. cruzi. Interestingly, we describe a hypothetical hybrid interactome of PSPs which reveals that T. cruzi secreted molecules may be down-regulating IL-17 whilst T. rangeli may enhance the production of IL-15. These results will pave the way for a better understanding of the pathophysiology of Chagas disease and may ultimately lead to the identification of molecular targets, such as key PSPs, that could be used to minimize the health outcomes of Chagas disease by modulating the immune response triggered by T. cruzi infection.
Collapse
Affiliation(s)
- Renata Watanabe Costa
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marina Ferreira Batista
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela Meneghelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ramon Oliveira Vidal
- The Berlin Institute for Medical Systems Biology-Max Delbrück Center for Molecular Medicine in the Helmholtz Association in Berlin, Berlin, Germany.,Laboratorio Nacional de Biociências (LNBio), Campinas, São Paulo, Brazil
| | - Carlos Alcides Nájera
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Clara Mendes
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela Augusta Andrade-Lima
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - José Franco da Silveira
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luciano Rodrigo Lopes
- Departamento de Informática em Saúde, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ludmila Rodrigues Pinto Ferreira
- RNA Systems Biology Lab (RSBL), Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernando Antoneli
- Departamento de Informática em Saúde, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Diana Bahia
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
22
|
Amezcua Vesely MC, Rodríguez C, Gruppi A, Acosta Rodríguez EV. Interleukin-17 mediated immunity during infections with Trypanosoma cruzi and other protozoans. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165706. [PMID: 31987839 PMCID: PMC7071987 DOI: 10.1016/j.bbadis.2020.165706] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/05/2019] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
Host resistance during infection with Trypanosoma cruzi, and other protozoans, is dependent on a balanced immune response. Robust immunity against these pathogens requires of the concerted action of many innate and adaptive cell populations including macrophages, neutrophils, dendritic cells, CD4+, and CD8+ T cells and B cells among others. Indeed, during most protozoan infections only a balanced production of inflammatory (TH1) and anti-inflammatory (TH2/regulatory) cytokines will allow the control of parasite spreading without compromising host tissue integrity. The description of TH17 cells, a novel effector helper T cell lineage that produced IL-17 as signature cytokine, prompted the revision of our knowledge about the mechanisms that mediate protection and immunopathology during protozoan infections. In this manuscript we discuss the general features of IL-17 mediated immune responses as well as the cellular sources, effector mechanisms and overall role of IL-17 in the immune response to T. cruzi and other protozoan infections.
Collapse
Affiliation(s)
- María Carolina Amezcua Vesely
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Constanza Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Eva Virginia Acosta Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
23
|
Sanchez Alberti A, Bivona AE, Matos MN, Cerny N, Schulze K, Weißmann S, Ebensen T, González G, Morales C, Cardoso AC, Cazorla SI, Guzmán CA, Malchiodi EL. Mucosal Heterologous Prime/Boost Vaccination Induces Polyfunctional Systemic Immunity, Improving Protection Against Trypanosoma cruzi. Front Immunol 2020; 11:128. [PMID: 32153562 PMCID: PMC7047160 DOI: 10.3389/fimmu.2020.00128] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 01/17/2020] [Indexed: 12/15/2022] Open
Abstract
There are several unmet needs in modern immunology. Among them, vaccines against parasitic diseases and chronic infections lead. Trypanosoma cruzi, the causative agent of Chagas disease, is an excellent example of a silent parasitic invasion that affects millions of people worldwide due to its progression into the symptomatic chronic phase of infection. In search for novel vaccine candidates, we have previously introduced Traspain, an engineered trivalent immunogen that was designed to address some of the known mechanisms of T. cruzi immune evasion. Here, we analyzed its performance in different DNA prime/protein boost protocols and characterized the systemic immune response associated with diverse levels of protection. Formulations that include a STING agonist, like c-di-AMP in the boost doses, were able to prime a Th1/Th17 immune response. Moreover, comparison between them showed that vaccines that were able to prime polyfunctional cell-mediated immunity at the CD4 and CD8 compartment enhanced protection levels in the murine model. These findings contribute to a better knowledge of the desired vaccine-elicited immunity against T. cruzi and promote the definition of a vaccine correlate of protection against the infection.
Collapse
Affiliation(s)
- Andrés Sanchez Alberti
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Augusto E Bivona
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marina N Matos
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Natacha Cerny
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sebastian Weißmann
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Germán González
- Departamento de Patología, Facultad de Medicina, Instituto de Fisiopatología Cardiovascular, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Celina Morales
- Departamento de Patología, Facultad de Medicina, Instituto de Fisiopatología Cardiovascular, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro C Cardoso
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvia I Cazorla
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Emilio L Malchiodi
- Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
24
|
Vásquez Velásquez C, Russomando G, Espínola EE, Sanchez Z, Mochizuki K, Roca Y, Revollo J, Guzman A, Quiroga B, Rios Morgan S, Vargas Ortiz R, Zambrana Ortega A, Espinoza E, Nishizawa JE, Kamel MG, Kikuchi M, Mizukami S, Na-Bangchang K, Tien Huy N, Hirayama K. IL-17A, a possible biomarker for the evaluation of treatment response in Trypanosoma cruzi infected children: A 12-months follow-up study in Bolivia. PLoS Negl Trop Dis 2019; 13:e0007715. [PMID: 31553732 PMCID: PMC6760767 DOI: 10.1371/journal.pntd.0007715] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 08/16/2019] [Indexed: 12/14/2022] Open
Abstract
Background The National Program for Chagas disease was implemented in Bolivia in 2006, and it greatly decreased the number of infections through vector control. Subsequently, a treatment regimen of benznidazole (BNZ) was started in seropositive school-age children living in certified vector control areas. Methods and findings We conducted a 12-month follow-up study and seven blood samples were taken during and after the treatment. Serology, conventional diagnostic PCR (cPCR) and quantitative Real-time PCR (qPCR) were performed. Plasma Th1/Th2/Th17 cytokines levels were also determined. Approximately 73 of 103 seropositive children complied with BNZ, with three interruptions due to side effects. To evaluate each individual’s treatment efficacy, the cPCR and qPCR values during the final 6 months of the follow-up period were observed. Among 57 children who completed follow-up, 6 individuals (11%) showed both cPCR(+) and qPCR(+) (non reactive), 24 (42%) cPCR(-) but qPCR(+) (ambiguous) and 27 (47%) cPCR(-) and qPCR(-) (reactive). Within 14 Th1/Th2/Th17 cytokines, IL-17A showed significantly higher levels in seropositive children before the treatment compared to age-matched seronegative children and significantly decreased to the normal level one-year after. Moreover, throughout the follow-up study, IL-17A levels were positively co-related to parasite counts detected by qPCR. At the 12 months’ time point, IL-17A levels of non-reactive subjects were significantly higher than either those of reactive or ambiguous subjects suggesting that IL-17A might be useful to determine the reactivity to BNZ treatment. Conclusions Plasma levels of IL-17A might be a bio-marker for detecting persistent infection of T. cruzi and its chronic inflammation. Chagas is a zoonosis endemic in 21 Latin American countries caused by T. cruzi. Results of common Benznidazole (BNZ) treatment vary by infection phase, treatment period, and dosage. In Bolivia, the national Chagas program controls vector distribution in different regions of the country. The program began BNZ treatment in school-age children from infestation-free endemic areas. Lack of information regarding follow-up and efficacy in children with recent chronic Chagas makes treatment failure difficult to detect in endemic areas. The present study aimed to estimate parasite DNA in blood through quantitative real-time and conventional PCR (qPCR, cPCR), and observe Th1/Th2/Th17 cytokine profiling during a 12-month follow-up in Bolivia school children. Results showed persistence of low, substantial amounts of T. cruzi DNA, and significantly higher IL-17A levels in the seropositive group before treatment than the seronegative group, which decreased to seronegative levels one year later. Of 57 treated, 6 showed cPCR positive results 6 months after treatment and were diagnosed as definitely non-reactive (10.5%). The six non-reactive individuals showed significantly higher levels of IL-17A at 12 months than residual reactive (cPCR negative, qPCR negative) and ambiguously reactive (cPCR negative, qPCR positive) groups, indicating that IL-17A might be a biomarker for non-reactive to BNZ.
Collapse
Affiliation(s)
- Clara Vásquez Velásquez
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, Japan
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Graciela Russomando
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Asunción, Asunción, Paraguay
| | - Emilio E. Espínola
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Asunción, Asunción, Paraguay
| | - Zunilda Sanchez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones en Ciencias de la Salud, Universidad Nacional de Asunción, Asunción, Paraguay
| | - Kota Mochizuki
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, Japan
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Yelin Roca
- Centro Nacional de Enfermedades Tropicales (CENETROP), Santa Cruz, Bolivia
| | - Jimmy Revollo
- Centro Nacional de Enfermedades Tropicales (CENETROP), Santa Cruz, Bolivia
| | - Angelica Guzman
- Centro Nacional de Enfermedades Tropicales (CENETROP), Santa Cruz, Bolivia
| | - Benjamín Quiroga
- Programa Departamental de Control de Chagas del Ministerio de Salud, Santa Cruz, Bolivia
| | - Susana Rios Morgan
- Programa Departamental de Control de Chagas del Ministerio de Salud, Santa Cruz, Bolivia
| | - Roberto Vargas Ortiz
- Programa Departamental de Control de Chagas del Ministerio de Salud, Santa Cruz, Bolivia
| | | | - Eida Espinoza
- Hospital Municipal Warnes "Nuestra Señora del Rosario", Santa Cruz, Bolivia
| | | | | | - Mihoko Kikuchi
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Shusaku Mizukami
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Kesara Na-Bangchang
- Chulabhorn International College of Medicine, Thammasat University, Pathumthani, Thailand
| | - Nguyen Tien Huy
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan
- Department of Clinical Product Development, Institute of Tropical Medicine (NEKKEN), School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki, Japan
- Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
25
|
Nagajyothi JF, Weiss LM. Advances in understanding the role of adipose tissue and mitochondrial oxidative stress in Trypanosoma cruzi infection. F1000Res 2019; 8. [PMID: 31354939 PMCID: PMC6652099 DOI: 10.12688/f1000research.19190.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/16/2019] [Indexed: 01/25/2023] Open
Abstract
Trypanosoma cruzi, the etiologic agent of Chagas disease, causes a latent infection that results in cardiomyopathy. Infection with this pathogen is a major socio-economic burden in areas of endemic infection throughout Latin America. The development of chagasic cardiomyopathy is dependent on the persistence of this parasite in host tissues. Pathogenesis of this cardiomyopathy is multifactorial and research indicates that it includes microvascular dysfunction, immune responses to host and parasite antigens, and various vasoactive and lipid mediators produced by both the host and parasite. It has been demonstrated that
T. cruzi persists in adipose tissue and uses fat as a nutritional niche in infected hosts. This chronic infection of adipose tissue plays an important role in the pathogenesis and persistence of this infection and involves mitochondrial stress responses as well as the production of various anti-inflammatory adipokines and pro-inflammatory cytokines by both white and brown adipose tissue. The changes in diet in endemic regions of infection have resulted in an epidemic of obesity that has significant implications for the pathogenesis of
T. cruzi infection and the development of chagasic cardiomyopathy in infected humans.
Collapse
Affiliation(s)
- Jyothi F Nagajyothi
- Department of Microbiology, Biochemistry and Molecular Genetics, Public Health Research Institute, New Jersey Medical School, 225 Warren Street, Newark, NJ, 07103, USA
| | - Louis M Weiss
- Departments of Pathology and Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Room 504 Forchheimer Building, Bronx, NY, 10461, USA
| |
Collapse
|
26
|
Medeiros NI, Gomes JAS. Cytometric Bead Array (CBA) for Measuring Cytokine Levels in Chagas Disease Patients. Methods Mol Biol 2019; 1955:309-314. [PMID: 30868537 DOI: 10.1007/978-1-4939-9148-8_23] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cytometric bead array (CBA) is a flow cytometry application that allows users to quantify multiple proteins simultaneously. Compared to other quantifier assays, as enzyme-linked immunosorbent assay (ELISA) and Western blot, CBA significantly reduces sample requirements and time to results. This technology allows for the design and creation of assays to measure a variety of analytes including inflammatory mediators, chemokines, immunoglobulin isotypes, intracellular signaling molecules, apoptotic mediators, adhesion molecules, and antibodies. Here we describe CBA steps to measure soluble cytokine levels in body fluids of infected patients by the protozoan Trypanosoma cruzi, morbidity known as Chagas disease.
Collapse
Affiliation(s)
- Nayara I Medeiros
- Cellular and Molecular Immunology, René Rachou Institute, Oswaldo Cruz Foundation, Belo Horizonte, Minas Gerais, Brazil
- Laboratory of Cell-Cell Interactions, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana A S Gomes
- Laboratory of Cell-Cell Interactions, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
27
|
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: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
28
|
Felizardo AA, Caldas IS, Mendonça AAS, Gonçalves RV, Tana FL, Almeida LA, Novaes RD. Impact of Trypanosoma cruzi infection on nitric oxide synthase and arginase expression and activity in young and elderly mice. Free Radic Biol Med 2018; 129:227-236. [PMID: 30248443 DOI: 10.1016/j.freeradbiomed.2018.09.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/21/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
Elderly organisms are more susceptible to infectious diseases. However, the impact of aging on antiparasitic mechanisms, especially the nitric oxide pathway, is poorly understood. Using an integrated in vivo and in vitro model, we compared the severity of Trypanosoma cruzi infection in young and elderly (8 or 72 weeks old) mice. Forty C57BL/6 mice were randomized into four groups: Y-inf, young infected; Yn-inf, young uninfected; A-inf, aged infected; An-inf, aged uninfected. Parasitemia was measured daily, and animals were euthanized after 15 days of infection. Trypanosoma cruzi-induced inflammatory processes were analyzed in blood and heart samples, as well as in bone marrow-derived macrophages (BMDMs) co-cultured with splenocytes isolated from young or elderly mice. Our results indicated upregulated IgG2b and IL-17 production in elderly animals, which was not sufficient to reduce parasitemia, parasitic load and myocarditis to levels observed in young animals. The higher susceptibility of elderly mice to T. cruzi infection was accompanied by reduced cardiac inducible nitric oxide synthase (iNOS) gene expression, nitric oxide (NO) and IFN-γ levels, as well as an antagonistic upregulation of arginase-1 expression and arginase activity. The same responses were observed when BMDMs co-cultured with splenocytes from elderly mice were stimulated with T. cruzi antigens. Our findings indicate that elderly mice were more susceptible to T. cruzi infection, which was potentially related to an attenuated response to antigenic stimulation, inhibition of iNOS gene expression and NO production, and antagonistic upregulation of arginase gene expression and activity, which created favorable conditions for heart parasitism and myocarditis development.
Collapse
Affiliation(s)
- Amanda A Felizardo
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Ivo S Caldas
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Andréa A S Mendonça
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-000, Minas Gerais, Brazil
| | - Fernanda L Tana
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Leonardo A Almeida
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Rômulo D Novaes
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil.
| |
Collapse
|
29
|
Upregulation of Cardiac IL-10 and Downregulation of IFN- γ in Balb/c IL-4 -/- in Acute Chagasic Myocarditis due to Colombian Strain of Trypanosoma cruzi. Mediators Inflamm 2018; 2018:3421897. [PMID: 30622430 PMCID: PMC6304210 DOI: 10.1155/2018/3421897] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
Inflammatory response in Chagas disease is related to parasite and host factors. However, immune system regulation has not been fully elucidated. Thus, this study is aimed at evaluating IL-4 influence on acute phase of Trypanosoma cruzi experimental infection through dosage of cytokine levels in cardiac homogenate of infected Balb/c WT and Balb/c IL-4−/− as well as its histopathological repercussions. For such purpose, mice were divided into two groups: an infected group with 100 forms of the Colombian strain and an uninfected group. After 21 days of infection, animals were euthanized and the blood, spleen, and heart were collected. The spleen was used to culture splenic cells in 48 h. Subsequently, cytokines TNF-α, IL-12p70, IL-10, IFN-γ, and IL-17 were measured in the blood, culture supernatant, and heart apex by ELISA. The base of the heart was used for histopathological analysis. From these analysis, infected Balb/c IL-4−/− mice showed milder inflammatory infiltrate compared to Balb/c WT, but without changes in nest density and collagen deposition. IL-4 absence culminated in lower cardiac tissue IFN-γ production, although it did not affect TNF-α expression in situ. It also decreased TNF-α systemic production and increased IL-10, both systemically and in situ. In addition, IL-4 absence did not influence IL-17 expression. Splenocytes of IL-4-deficient mice produced higher amounts of IFN-γ, TNF-α, and IL-17 and lower amounts of IL-10. Thus, IL-4 absence in acute phase of experimental infection with T. cruzi Colombian strain reduces myocarditis due to lower IFN-γ production and greater IL-10 production in situ and this pattern is not influenced by splenocyte general repertoire.
Collapse
|
30
|
Pereira FB, Dutra WO, Gollob KJ, Reis EA, Oliveira ALGD, Rocha MODC, Menezes CADS. Vasoactive intestinal peptide degradation might influence Interleukin-17 expression in cardiac chagasic patients. Rev Inst Med Trop Sao Paulo 2018; 60:e57. [PMID: 30365640 PMCID: PMC6199123 DOI: 10.1590/s1678-9946201860057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/14/2018] [Indexed: 12/03/2022] Open
Abstract
The vasoactive intestinal peptide (VIP) expression is lower in cardiac chagasic patients and is related to worse cardiac function. The reduction of VIP in patients with Chagas disease may be a result of its enhanced degradation. To test this hypothesis, the tryptase and chymase expression was evaluated. We also related VIP levels with interleukin-17 (IL-17) expression since VIP may modulate IL-17 production. Plasma levels of chymase were higher in chagasic patients. Conversely, VIP/chymase and VIP/tryptase ratios were lower in chagasic patients when compared to non-infected individuals. Besides, the VIP/chymase ratio was lower in chagasic cardiac patients in comparison with the indeterminate group. A positive correlation between tryptase and chymase levels was observed in chagasic cardiac patients. In relation to IL-17, we observed a higher expression of this cytokine in the cardiac form of the disease than in the indeterminate form. IL-17/VIP ratio was higher in the cardiac form in comparison with non-infected or indeterminate form. These results suggest that the low levels of VIP observed in chagasic patients could be due to an increased production of chymase and/or to the additive effect of the interaction between chymase and tryptase in the cardiac form. Moreover, the decreased VIP expression may contribute to the increase of IL-17 in chagasic cardiac patients.
Collapse
Affiliation(s)
| | - Walderez O. Dutra
- Universidade Federal de Minas Gerais, Brazil; Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, Brazil
| | - Kenneth J. Gollob
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais, Brazil; A.C. Camargo Cancer Center, Brazil
| | | | | | | | | |
Collapse
|
31
|
Acevedo GR, Girard MC, Gómez KA. The Unsolved Jigsaw Puzzle of the Immune Response in Chagas Disease. Front Immunol 2018; 9:1929. [PMID: 30197647 PMCID: PMC6117404 DOI: 10.3389/fimmu.2018.01929] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Trypanosoma cruzi interacts with the different arms of the innate and adaptive host's immune response in a very complex and flowery manner. The history of host-parasite co-evolution has provided this protozoan with means of resisting, escaping or subverting the mechanisms of immunity and establishing a chronic infection. Despite many decades of research on the subject, the infection remains incurable, and the factors that steer chronic Chagas disease from an asymptomatic state to clinical onset are still unclear. As the relationship between T. cruzi and the host immune system is intricate, so is the amount and diversity of scientific knowledge on the matter. Many of the mechanisms of immunity are fairly well understood, but unveiling the factors that lead each of these to success or failure, within the coordinated response as a whole, requires further research. The intention behind this Review is to compile the available information on the different aspects of the immune response, with an emphasis on those phenomena that have been studied and confirmed in the human host. For ease of comprehension, it has been subdivided in sections that cover the main humoral and cell-mediated components involved therein. However, we also intend to underline that these elements are not independent, but function intimately and concertedly. Here, we summarize years of investigation carried out to unravel the puzzling interplay between the host and the parasite.
Collapse
Affiliation(s)
| | | | - Karina A. Gómez
- Laboratorio de Inmunología de las Infecciones por Tripanosomátidos, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
32
|
De Alba-Alvarado M, Salazar-Schettino PM, Jiménez-Álvarez L, Cabrera-Bravo M, García-Sancho C, Zenteno E, Vazquez-Antona C, Cruz-Lagunas A, Zúñiga J, Bucio-Torres MI. Th-17 cytokines are associated with severity of Trypanosoma cruzi chronic infection in pediatric patients from endemic areas of Mexico. Acta Trop 2018; 178:134-141. [PMID: 29180164 DOI: 10.1016/j.actatropica.2017.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 11/07/2017] [Accepted: 11/20/2017] [Indexed: 01/07/2023]
Abstract
In Chagas disease the clinical, acute and chronic manifestations are the result of the interaction between the parasite and the host factors. The balance between inflammatory and anti-inflammatory immune responses is essential for the increase or resolution of the manifestations in individuals infected with T. cruzi. To identify if children with chronic Chagas disease and heart injury is related with non-regulated Th1, Th2 and Th17 responses. We included 31 children with T. cruzi confirmed chronic infection from endemic areas of Mexico. Subsequently, they were separated according to their ECHO and ECG results into three groups according to the severity of cardiac involvement. Circulating Th1, Th2 and Th17 cytokine profiles were performed by Luminex assays and the results were analyzed by bivariate and multivariable analysis. Patients were classified in asymptomatic chronic (group 1, N=12); individuals with IRBBB in ECG and incipient lesions in ECHO (Group 2, N=8) and Patients with severe chronic symptomatic disease (Group 3, N=11). The analysis of immune mediators revealed that patients with severe cardiac manifestations had significant higher levels (p <0.05) of Th17 related cytokines including IL-17 and IL-6 as well as IFN-γ and IL-2. Also patients with severe cardiomyopathy exhibit increased levels of IL-13 (p <0.05) after multivariate analysis. High levels of Th17 related cytokines including IL-17, IFN-γ, IL-6 and IL-2 and pro-fibrotic factors such as IL-13 could be associated to the severity of cardiac involvement in children with chronic T. cruzi infection. These cytokines could be useful as indicators for the early identification of cardiac damage associated to the T. cruzi infection.
Collapse
Affiliation(s)
- Mariana De Alba-Alvarado
- Department of Microbiology and Parasitology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Paz María Salazar-Schettino
- Department of Microbiology and Parasitology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Luis Jiménez-Álvarez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Margarita Cabrera-Bravo
- Department of Microbiology and Parasitology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Cecilia García-Sancho
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Edgar Zenteno
- Department of Microbiology and Parasitology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Clara Vazquez-Antona
- Department of Microbiology and Parasitology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Martha Irene Bucio-Torres
- Department of Microbiology and Parasitology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico.
| |
Collapse
|
33
|
Sanmarco LM, Eberhardt N, Ponce NE, Cano RC, Bonacci G, Aoki MP. New Insights into the Immunobiology of Mononuclear Phagocytic Cells and Their Relevance to the Pathogenesis of Cardiovascular Diseases. Front Immunol 2018; 8:1921. [PMID: 29375564 PMCID: PMC5767236 DOI: 10.3389/fimmu.2017.01921] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/14/2017] [Indexed: 12/18/2022] Open
Abstract
Macrophages are the primary immune cells that reside within the myocardium, suggesting that these mononuclear phagocytes are essential in the orchestration of cardiac immunity and homeostasis. Independent of the nature of the injury, the heart triggers leukocyte activation and recruitment. However, inflammation is harmful to this vital terminally differentiated organ with extremely poor regenerative capacity. As such, cardiac tissue has evolved particular strategies to increase the stress tolerance and minimize the impact of inflammation. In this sense, growing evidences show that mononuclear phagocytic cells are particularly dynamic during cardiac inflammation or infection and would actively participate in tissue repair and functional recovery. They respond to soluble mediators such as metabolites or cytokines, which play central roles in the timing of the intrinsic cardiac stress response. During myocardial infarction two distinct phases of monocyte influx have been identified. Upon infarction, the heart modulates its chemokine expression profile that sequentially and actively recruits inflammatory monocytes, first, and healing monocytes, later. In the same way, a sudden switch from inflammatory macrophages (with microbicidal effectors) toward anti-inflammatory macrophages occurs within the myocardium very shortly after infection with Trypanosoma cruzi, the causal agent of Chagas cardiomyopathy. While in sterile injury, healing response is necessary to stop tissue damage; during an intracellular infection, the anti-inflammatory milieu in infected hearts would promote microbial persistence. The balance of mononuclear phagocytic cells seems to be also dynamic in atherosclerosis influencing plaque initiation and fate. This review summarizes the participation of mononuclear phagocyte system in cardiovascular diseases, keeping in mind that the immune system evolved to promote the reestablishment of tissue homeostasis following infection/injury, and that the effects of different mediators could modulate the magnitude and quality of the immune response. The knowledge of the effects triggered by diverse mediators would serve to identify new therapeutic targets in different cardiovascular pathologies.
Collapse
Affiliation(s)
- Liliana Maria Sanmarco
- 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 Tecnológicas (CONICET), Córdoba, Argentina
| | - Natalia Eberhardt
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Córdoba, Argentina
| | - Nicolás Eric Ponce
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Roxana Carolina Cano
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Universidad Católica de Córdoba, Unidad Asociada Área Ciencias Agrarias, Ingeniería, Ciencias Biológicas y de la Salud, Facultad de Ciencias Químicas, Córdoba, Argentina
| | - Gustavo Bonacci
- 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 Tecnológicas (CONICET), Córdoba, Argentina
| | - Maria Pilar Aoki
- 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 Tecnológicas (CONICET), Córdoba, Argentina
| |
Collapse
|
34
|
Medeiros NI, Gomes JAS, Correa-Oliveira R. Synergic and antagonistic relationship between MMP-2 and MMP-9 with fibrosis and inflammation in Chagas' cardiomyopathy. Parasite Immunol 2017; 39. [PMID: 28543409 DOI: 10.1111/pim.12446] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022]
Abstract
Cardiomyopathy is the most important clinical manifestation in the chronic phase of Chagas' disease because of its frequency, severity and impact on morbidity and mortality. The extracellular matrix degradation during cardiac remodeling in Trypanosoma cruzi infection is driven by matrix metalloproteinases (MMPs), primarily the MMP-2 and MMP-9 gelatinases. MMPs also regulate some molecules related to inflammation, such as growth factors, cytokines and chemokines. The involvement of MMP-2 and MMP-9 is not yet fully understood in Chagas' disease. It has been proposed that the gelatinases may have opposite effect on inflammation/regulation and cardiac remodeling. MMP-2 would participate in regulation, offering a protective role for cardiac damage in asymptomatic patients and would be a good marker for the initiation of changes in the heart. On the other hand, MMP-9 can be used as a marker for serious changes on the heart and would be associated with inflammation and fibrosis. Here, we consolidate all characteristics involving MMP-2 and MMP-9 in Chagas' disease based on current studies to clarify their participation on the inflammation/regulation and fibrosis, and the synergistic or antagonistic role between them.
Collapse
Affiliation(s)
- N I Medeiros
- Imunologia Celular e Molecular, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil.,Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - J A S Gomes
- Departamento de Morfologia, Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - R Correa-Oliveira
- Imunologia Celular e Molecular, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| |
Collapse
|