1
|
Khan AA, Taylor MC, Fortes Francisco A, Jayawardhana S, Atherton RL, Olmo F, Lewis MD, Kelly JM. Animal models for exploring Chagas disease pathogenesis and supporting drug discovery. Clin Microbiol Rev 2024; 37:e0015523. [PMID: 39545730 PMCID: PMC11629624 DOI: 10.1128/cmr.00155-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
SUMMARYInfections with the parasitic protozoan Trypanosoma cruzi cause Chagas disease, which results in serious cardiac and/or digestive pathology in 30%-40% of individuals. However, symptomatic disease can take decades to become apparent, and there is a broad spectrum of possible outcomes. The complex and long-term nature of this infection places a major constraint on the scope for experimental studies in humans. Accordingly, predictive animal models have been a mainstay of Chagas disease research. The resulting data have made major contributions to our understanding of parasite biology, immune responses, and disease pathogenesis and have provided a platform that informs and facilitates the global drug discovery effort. Here, we provide an overview of available animal models and illustrate how they have had a key impact across the field.
Collapse
Affiliation(s)
- Archie A. Khan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Richard L. Atherton
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
2
|
Sabino EC, Nunes MCP, Blum J, Molina I, Ribeiro ALP. Cardiac involvement in Chagas disease and African trypanosomiasis. Nat Rev Cardiol 2024; 21:865-879. [PMID: 39009679 DOI: 10.1038/s41569-024-01057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 07/17/2024]
Abstract
Trypanosomiases are diseases caused by various species of protozoan parasite in the genus Trypanosoma, each presenting with distinct clinical manifestations and prognoses. Infections can affect multiple organs, with Trypanosoma cruzi predominantly affecting the heart and digestive system, leading to American trypanosomiasis or Chagas disease, and Trypanosoma brucei primarily causing a disease of the central nervous system known as human African trypanosomiasis or sleeping sickness. In this Review, we discuss the effects of these infections on the heart, with particular emphasis on Chagas disease, which continues to be a leading cause of cardiomyopathy in Latin America. The epidemiology of Chagas disease has changed substantially since 1990 owing to the emigration of over 30 million Latin American citizens, primarily to Europe and the USA. This movement of people has led to the global dissemination of individuals infected with T. cruzi. Therefore, cardiologists worldwide must familiarize themselves with Chagas disease and the severe, chronic manifestation - Chagas cardiomyopathy - because of the expanded prevalence of this disease beyond traditional endemic regions.
Collapse
Affiliation(s)
- Ester Cerdeira Sabino
- Department of Pathology, Instituto de Medicina Tropical da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, Brazil.
| | - Maria Carmo P Nunes
- Hospital das Clínicas and Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Johannes Blum
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Israel Molina
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, PROSICS Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Luiz P Ribeiro
- Hospital das Clínicas and Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
3
|
Veluswami K, Rao S, Aggarwal S, Mani S, Balasubramanian A. Unraveling the Missing Pieces: Exploring the Gaps in Understanding Chagas Cardiomyopathy. Cureus 2024; 16:e66955. [PMID: 39280489 PMCID: PMC11401617 DOI: 10.7759/cureus.66955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2024] [Indexed: 09/18/2024] Open
Abstract
Chagas cardiomyopathy affects a considerable number of patients infected with the protozoan Trypanosoma cruzi (T. cruzi) and remains one of the most neglected tropical diseases despite being a significant contributor to morbidity and mortality in both endemic regions of Latin America and non-endemic countries like the United States. Since its discovery almost a century ago, knowledge gaps still exist in the mechanisms involved in the pathogenesis of Chagas cardiomyopathy, and numerous challenges exist in its diagnosis and treatment. This article reviews the main pathogenetic mechanisms involved in the progression of Chagas cardiomyopathy, which has been proposed as a result of years of research. It also emphasizes the challenges involved in the diagnosis of the asymptomatic indeterminate phase and has focused on several diagnostic techniques, including echocardiography, electrocardiogram (ECG), magnetic resonance imaging (MRI), and nuclear imaging in diagnosing symptomatic Chagas cardiomyopathy. In this article, we have also provided a brief overview of the current treatment of Chagas cardiomyopathy, which is not etiology-specific but instead derived from the knowledge acquired from the treatment of other cardiomyopathies.
Collapse
Affiliation(s)
| | - Sudipta Rao
- Internal Medicine, JSS Medical College, Mysore, IND
| | | | - Sweatha Mani
- Internal Medicine, K.A.P. Viswanatham Government Medical College, Tiruchirappalli, IND
| | | |
Collapse
|
4
|
Yao Z, Liang M, Zhu S. Infectious factors in myocarditis: a comprehensive review of common and rare pathogens. Egypt Heart J 2024; 76:64. [PMID: 38789885 PMCID: PMC11126555 DOI: 10.1186/s43044-024-00493-3] [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: 03/07/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND Myocarditis is a significant health threat today, with infectious agents being the most common cause. Accurate diagnosis of the etiology of infectious myocarditis is crucial for effective treatment. MAIN BODY Infectious myocarditis can be caused by viruses, prokaryotes, parasites, and fungi. Viral infections are typically the primary cause. However, some rare opportunistic pathogens can also damage heart muscle cells in patients with immunodeficiencies, neoplasms and those who have undergone heart surgery. CONCLUSIONS This article reviews research on common and rare pathogens of infectious myocarditis, emphasizing the complexity of its etiology, with the aim of helping clinicians make an accurate diagnosis of infectious myocarditis.
Collapse
Affiliation(s)
- Zongjie Yao
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qindao, China.
| | - Mingjun Liang
- Department of Intensive Care Medicine, Shanghai Six People's Hospital Affilicated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Simin Zhu
- Wuhan Third Hospital-Tongren Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
5
|
do Vale INPC, Almeida GG, Rimkute I, Liechti T, de Araújo FF, dos Santos LI, Henriques PM, Rocha MODC, Elói-Santos SM, Martins−Filho OA, Roederer M, Sher A, Jankovic D, Teixeira−Carvalho A, Antonelli LRDV. Signatures of CD4 + T and B cells are associated with distinct stages of chronic chagasic cardiomyopathy. Front Immunol 2024; 15:1385850. [PMID: 38726014 PMCID: PMC11079136 DOI: 10.3389/fimmu.2024.1385850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 03/27/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Chagas disease is a neglected parasitic disease caused by Trypanosoma cruzi. While most patients are asymptomatic, around 30% develop Chronic Chagasic Cardiomyopathy (CCC). Methods Here, we employed high-dimensional flow cytometry to analyze CD4+ T and B cell compartments in patients during the chronic phase of Chagas disease, presenting the asymptomatic and mild or moderate/severe cardiac clinical forms. Results Effector CD27-CD4+ T cells were expanded in both CCC groups, and only mild CCC patients showed higher frequencies of effector memory and T follicular helper (Tfh) cells than healthy donors (CTL) and asymptomatic patients. Unsupervised analysis confirmed these findings and further revealed the expansion of a specific subpopulation composed of Tfh, transitional, and central memory CD4+ T cells bearing a phenotype associated with strong activation, differentiation, and exhaustion in patients with mild but not moderate/severe CCC. In contrast, patients with mild and moderate/severe CCC had lower frequencies of CD4+ T cells expressing lower levels of activation markers, suggesting resting status, than CTL. Regarding the B cell compartment, no alterations were found in naïve CD21-, memory cells expressing IgM or IgD, marginal zone, and plasma cells in patients with Chagas disease. However, expansion of class-switched activated and atypical memory B cells was observed in all clinical forms, and more substantially in mild CCC patients. Discussion Taken together, our results showed that T. cruzi infection triggers changes in CD4+ T and B cell compartments that are more pronounced in the mild CCC clinical form, suggesting an orchestrated cellular communication during Chagas disease. Conclusion Overall, these findings reinforce the heterogeneity and complexity of the immune response in patients with chronic Chagas disease and may provide new insights into disease pathology and potential markers to guide clinical decisions.
Collapse
Affiliation(s)
- Isabela Natália Pascoal Campos do Vale
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
- Integrated Research Group in Biomarkers, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
| | - Gregório Guilherme Almeida
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
| | - Inga Rimkute
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Thomas Liechti
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Fernanda Fortes de Araújo
- Integrated Research Group in Biomarkers, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
| | - Luara Isabela dos Santos
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
- Departament of Basic Science, Faculty of Medical Sciences of Minas Gerais, Belo Horizonte, Brazil
| | - Priscilla Miranda Henriques
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
| | - Manoel Otávio da Costa Rocha
- Department of Clinical Medicine, Postgraduate Program in Infectious Diseases and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Silvana Maria Elói-Santos
- Integrated Research Group in Biomarkers, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
- Department of Complementary Propedeutics, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Olindo Assis Martins−Filho
- Integrated Research Group in Biomarkers, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Andréa Teixeira−Carvalho
- Integrated Research Group in Biomarkers, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
| | - Lis Ribeiro do Valle Antonelli
- Biology and Immunology of Infectious and Parasitic Diseases Group, René Rachou Institute, Oswaldo Cruz Foundation-FIOCRUZ, Belo Horizonte, Brazil
| |
Collapse
|
6
|
Madeira RP, Meneghetti P, Lozano N, Namiyama GM, Pereira-Chioccola VL, Torrecilhas AC. Exploring Peripheral Blood-Derived Extracellular Vesicles as Biomarkers: Implications for Chronic Chagas Disease with Viral Infection or Transplantation. Microorganisms 2024; 12:116. [PMID: 38257943 PMCID: PMC10818975 DOI: 10.3390/microorganisms12010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer envelopes that encapsulate cell-specific cargo, rendering them promising biomarkers for diverse diseases. Chagas disease, caused by the parasite Trypanosoma cruzi, poses a significant global health burden, transcending its initial epicenter in Latin America to affect individuals in Europe, Asia, and North America. In this study, we aimed to characterize circulating EVs derived from patients with chronic Chagas disease (CCD) experiencing a reactivation of acute symptoms. Blood samples collected in EDTA were processed to isolate plasma and subsequently subjected to ultracentrifugation for particle isolation and purification. The EVs were characterized using a nanoparticle tracking analysis and enzyme-linked immunosorbent assay (ELISA). Our findings revealed distinctive differences in the size, concentration, and composition of EVs between immunosuppressed patients and those with CCD. Importantly, these EVs play a critical role in the pathophysiology of Chagas disease and demonstrate significant potential as biomarkers in the chronic phase of the disease. Overall, our findings support the potential utility of the CL-ELISA assay as a specific sensitive tool for detecting circulating EVs in chronic Chagasic patients, particularly those with recurrent infection following an immunosuppressive treatment or with concurrent HIV and Chagas disease. Further investigations are warranted to identify and validate the specific antigens or biomarkers responsible for the observed reactivity in these patient groups, which may have implications for diagnosis, the monitoring of treatment, and prognosis.
Collapse
Affiliation(s)
- Rafael Pedro Madeira
- Disciplina de Infectologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (R.P.M.); (N.L.)
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema 09913-030, Brazil;
| | - Paula Meneghetti
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema 09913-030, Brazil;
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil
| | - Nicholy Lozano
- Disciplina de Infectologia, Departamento de Medicina, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (R.P.M.); (N.L.)
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema 09913-030, Brazil;
| | - Gislene M. Namiyama
- Electron Microscopy Laboratory, Adolfo Lutz Institute, São Paulo 01246-900, Brazil;
| | - Vera Lucia Pereira-Chioccola
- Laboratório de Biologia Molecular de Fungos e Parasitas, Centro de Parasitologia e Micologia, Instituto Adolfo Lutz, São Paulo 01246-000, Brazil
| | - Ana Claudia Torrecilhas
- Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema 09913-030, Brazil;
| |
Collapse
|
7
|
Andrade MV, Noronha KVMDS, de Souza A, Motta-Santos AS, Braga PEF, Bracarense H, de Miranda MCC, Nascimento BR, Molina I, Martins-Melo FR, Perel P, Geissbühler Y, Quijano M, Machado IE, Ribeiro ALP. The economic burden of Chagas disease: A systematic review. PLoS Negl Trop Dis 2023; 17:e0011757. [PMID: 37992061 DOI: 10.1371/journal.pntd.0011757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/06/2023] [Accepted: 10/29/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Chagas disease (CD) is a neglected disease affecting millions worldwide, yet little is known about its economic burden. This systematic review is part of RAISE project, a broader study that aims to estimate the global prevalence, mortality, and health and economic burden attributable to chronic CD and Chronic Chagas cardiomyopathy. The objective of this study was to assess the main costs associated with the treatment of CD in both endemic and non-endemic countries. METHODS An electronic search of the Medline, Lilacs, and Embase databases was conducted until 31st, 2022, to identify and select economic studies that evaluated treatment costs of CD. No restrictions on place or language were made. Complete or partial economic analyses were included. RESULTS Fifteen studies were included, with two-thirds referring to endemic countries. The most commonly investigated cost components were inpatient care, exams, surgeries, consultation, drugs, and pacemakers. However, significant heterogeneity in the estimation methods and presentation of data was observed, highlighting the absence of standardization in the measurement methods and cost components. The most common component analyzed using the same metric was hospitalization. The mean annual hospital cost per patient ranges from $25.47 purchasing power parity US dollars (PPP-USD) to $18,823.74 PPP-USD, and the median value was $324.44 PPP-USD. The lifetime hospital cost per patient varies from $209,44 PPP-USD for general care to $14,351.68 PPP-USD for patients with heart failure. DISCUSSION Despite the limitations of the included studies, this study is the first systematic review of the costs of CD treatment. The findings underscore the importance of standardizing the measurement methods and cost components for estimating the economic burden of CD and improving the comparability of cost components magnitude and cost composition analysis. Finally, assessing the economic burden is essential for public policies designed to eliminate CD, given the continued neglect of this disease.
Collapse
Affiliation(s)
- Mônica Viegas Andrade
- Department of Economics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Aline de Souza
- Faculty of Economic Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - André Soares Motta-Santos
- Center for Health Technology Assessment, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Center for Development and Regional Planning, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Henrique Bracarense
- Faculty of Economic Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Bruno Ramos Nascimento
- Department of Internal Medicine, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Telehealth Center and Cardiology Service, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Israel Molina
- International Health Unit Vall d'Hebron-Drassanes, Infectious Diseases Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | | | | | - Isis Eloah Machado
- Department of Family Medicine, Mental and Collective Health, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Antônio Luiz Pinho Ribeiro
- Department of Internal Medicine, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Telehealth Center and Cardiology Service, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
8
|
Hideko Tatakihara VL, Malvezi AD, Pereira RS, Lucchetti BFC, Dos Santos LF, Cecchini R, Yamauchi LM, Yamada-Ogatta SF, Miranda KM, Verri WA, Martins-Pinge MC, Pinge-Filho P. The Therapeutic Potential of Angeli's Salt in Mitigating Acute Trypanosoma cruzi Infection in Mice. Pathogens 2023; 12:1063. [PMID: 37624023 PMCID: PMC10458646 DOI: 10.3390/pathogens12081063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023] Open
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi, is a neglected tropical disease prevalent in Latin America. Infected patients are treated to eliminate the parasite, reduce the cardiomyopathy risk, and interrupt the disease transmission cycle. The World Health Organization recognizes benznidazole (BZ) and nifurtimox as effective drugs for CD treatment. In the chronic phase, both drugs have low cure rates and serious side effects. T. cruzi infection causes intense tissue inflammation that controls parasite proliferation and CD evolution. Compounds that liberate nitric oxide (NO) (NO donors) have been used as anti-T. cruzi therapeutics. Currently, there is no evidence that nitroxyl (HNO) affects T. cruzi infection outcomes. This study investigated the effects of the HNO donor Angeli's salt (AS) on C57BL/6 mice infected with T. cruzi (Y strain, 5 × 103 trypomastigotes, intraperitoneally). AS reduced the number of parasites in the bloodstream and heart nests and increased the protective antioxidant capacity of erythrocytes in infected animals, reducing disease severity. Furthermore, in vitro experiments showed that AS treatment reduced parasite uptake and trypomastigote release by macrophages. Taken together, these findings from the murine model and in vitro testing suggest that AS could be a promising therapy for CD.
Collapse
Affiliation(s)
- Vera Lúcia Hideko Tatakihara
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil (L.F.D.S.)
| | - Aparecida Donizette Malvezi
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil (L.F.D.S.)
| | - Rito Santo Pereira
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil (L.F.D.S.)
| | - Bruno Fernando Cruz Lucchetti
- Laboratório de Fisiologia e Fisiopatologia Cardiovascular, Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil
- Departamento de Fisioterapia, Centro Universitário do Vale do Araguaia, Barra do Garças 78603-209, Mato Grosso, Brazil
| | - Lucas Felipe Dos Santos
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil (L.F.D.S.)
| | - Rubens Cecchini
- Laboratório de Fisiopatologia e Radicais Livres, Departamento de Imunologia, Parasitologia e Patologia Geral, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil
| | - Lucy Megumi Yamauchi
- Laboratório de Biologia Molecular de Microrganismos, Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil
| | - Sueli Fumie Yamada-Ogatta
- Laboratório de Biologia Molecular de Microrganismos, Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil
| | - Katrina M. Miranda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA
| | - Waldiceu A. Verri
- Laboratório de Pesquisa em Dor, Inflamação, Neuropatia e Câncer, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil
| | - Marli Cardoso Martins-Pinge
- Departamento de Fisioterapia, Centro Universitário do Vale do Araguaia, Barra do Garças 78603-209, Mato Grosso, Brazil
| | - Phileno Pinge-Filho
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Paraná, Brazil (L.F.D.S.)
| |
Collapse
|
9
|
Lemos JM, Brito da Silva MF, Dos Santos Carvalho AM, Vicente Gil HP, Fiaia Costa VA, Andrade CH, Braga RC, Grellier P, Muratov EN, Charneau S, Moreira-Filho JT, Dourado Bastos IM, Neves BJ. Multitask learning-driven identification of novel antitrypanosomal compounds. Future Med Chem 2023; 15:1449-1467. [PMID: 37701989 DOI: 10.4155/fmc-2023-0074] [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] [Indexed: 09/14/2023] Open
Abstract
Background: Chagas disease and human African trypanosomiasis cause substantial death and morbidity, particularly in low- and middle-income countries, making the need for novel drugs urgent. Methodology & results: Therefore, an explainable multitask pipeline to profile the activity of compounds against three trypanosomes (Trypanosoma brucei brucei, Trypanosoma brucei rhodesiense and Trypanosoma cruzi) were created. These models successfully discovered four new experimental hits (LC-3, LC-4, LC-6 and LC-15). Among them, LC-6 showed promising results, with IC50 values ranging 0.01-0.072 μM and selectivity indices >10,000. Conclusion: These results demonstrate that the multitask protocol offers predictivity and interpretability in the virtual screening of new antitrypanosomal compounds and has the potential to improve hit rates in Chagas and human African trypanosomiasis projects.
Collapse
Affiliation(s)
- Jade Milhomem Lemos
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Meryck Felipe Brito da Silva
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Alexandra Maria Dos Santos Carvalho
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, 70910-900, DF, Brazil
| | - Henric Pietro Vicente Gil
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Vinícius Alexandre Fiaia Costa
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| | - Carolina Horta Andrade
- LabMol - Laboratory for Molecular Modeling & Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, 74605-170, GO, Brazil
| | - Rodolpho Campos Braga
- InsilicAll Ltda, Av. Eng. Luis Carlos Berrini,1748 - Itaim Bibi, 04571-010, Sao Paulo, SP, Brazil
| | - Philippe Grellier
- UMR 7245 Molécules de Communication et Adaptation des Micro-organismes, Muséum National d'Histoire Naturelle, Équipe Parasites et Protistes Libres, Paris, 0575231, France
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, 27599, NC, USA
- Department of Pharmaceutical Sciences, Federal University of Paraiba, Joao Pessoa, 58059-900, PB, Brazil
| | - Sébastien Charneau
- Department of Cell Biology, Laboratory of Biochemistry & Protein Chemistry, Institute of Biological Sciences, University of Brasilia, Brasilia, 70910-900, DF, Brazil
| | - José Teófilo Moreira-Filho
- LabMol - Laboratory for Molecular Modeling & Drug Design, Faculty of Pharmacy, Federal University of Goiás, Goiânia, 74605-170, GO, Brazil
| | - Izabela Marques Dourado Bastos
- Pathogen-Host Interface Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, 70910-900, DF, Brazil
| | - Bruno Junior Neves
- LabChem - Laboratory of Cheminformatics, Faculty of Pharmacy, Federal University of Goiás, Goiânia,74605-170, GO, Brazil
| |
Collapse
|
10
|
Król G, Fortunka K, Majchrzak M, Piktel E, Paprocka P, Mańkowska A, Lesiak A, Karasiński M, Strzelecka A, Durnaś B, Bucki R. Metallic Nanoparticles and Core-Shell Nanosystems in the Treatment, Diagnosis, and Prevention of Parasitic Diseases. Pathogens 2023; 12:838. [PMID: 37375528 DOI: 10.3390/pathogens12060838] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The usage of nanotechnology in the fight against parasitic diseases is in the early stages of development, but it brings hopes that this new field will provide a solution to target the early stages of parasitosis, compensate for the lack of vaccines for most parasitic diseases, and also provide new treatment options for diseases in which parasites show increased resistance to current drugs. The huge physicochemical diversity of nanomaterials developed so far, mainly for antibacterial and anti-cancer therapies, requires additional studies to determine their antiparasitic potential. When designing metallic nanoparticles (MeNPs) and specific nanosystems, such as complexes of MeNPs, with the shell of attached drugs, several physicochemical properties need to be considered. The most important are: size, shape, surface charge, type of surfactants that control their dispersion, and shell molecules that should assure specific molecular interaction with targeted molecules of parasites' cells. Therefore, it can be expected that the development of antiparasitic drugs using strategies provided by nanotechnology and the use of nanomaterials for diagnostic purposes will soon provide new and effective methods of antiparasitic therapy and effective diagnostic tools that will improve the prevention and reduce the morbidity and mortality caused by these diseases.
Collapse
Affiliation(s)
- Grzegorz Król
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Kamila Fortunka
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Michał Majchrzak
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Białystok, Mickiewicza 2B, 15-222 Białystok, Poland
| | - Paulina Paprocka
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Angelika Mańkowska
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Agata Lesiak
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Maciej Karasiński
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland
| | - Agnieszka Strzelecka
- Department of Public Health , Institute of Health Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Bonita Durnaś
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Robert Bucki
- Department of Microbiology and Immunology, Institute of Medical Science, Collegium Medicum, Jan Kochanowski University, IX Wieków Kielc 19A, 25-317 Kielce, Poland
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland
| |
Collapse
|
11
|
Cevey ÁC, Pieralisi AV, Donato M, Rada J, Gelpi RJ, Mirkin GA, Goren NB, Penas FN. Macrophages Mediate Healing Properties of Fenofibrate in Experimental Chagasic Cardiomyopathy. ACS Infect Dis 2023; 9:213-220. [PMID: 36661566 DOI: 10.1021/acsinfecdis.2c00535] [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: 01/21/2023]
Abstract
Chronic cardiomyopathy is one of the most relevant outcomes of Chagas disease associated with parasite persistence and exacerbated inflammatory response. Fenofibrate, a third generation fibric acid derivative and peroxisome proliferator-activated receptor-α ligand, is involved in the regulation of inflammatory response. However, the participation of macrophages in this scenario has not been elucidated. Here we show, for the first time, that macrophages play a fundamental role in the fenofibrate-mediated modulation of heart pro-inflammatory response and fibrosis caused by the infection with Trypanosoma cruzi. Furthermore, macrophages are required for fenofibrate to improve the loss of ventricular function and this restoration correlates with an anti-inflammatory microenvironment. Understanding the contributions of macrophages to the healing properties of fenofibrate reinforces its potential use as a therapeutic drug, with the aim of helping to solve a public health problem, such as chronic Chagas disease.
Collapse
Affiliation(s)
- Ágata Carolina Cevey
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Azul Victoria Pieralisi
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Martín Donato
- Facultad de Medicina, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Jimena Rada
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Ricardo Jorge Gelpi
- Facultad de Medicina, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Gerardo Ariel Mirkin
- Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM). Facultad de Medicina, CONICET - Universidad de Buenos Aires. Buenos Aires C1121ABG, Argentina
| | - Nora Beatriz Goren
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| | - Federico Nicolás Penas
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS). Facultad de Medicina, CONICET - Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
12
|
Puerta CJ, Cuellar A, Lasso P, Mateus J, Gonzalez JM. Trypanosoma cruzi-specific CD8 + T cells and other immunological hallmarks in chronic Chagas cardiomyopathy: Two decades of research. Front Cell Infect Microbiol 2023; 12:1075717. [PMID: 36683674 PMCID: PMC9846209 DOI: 10.3389/fcimb.2022.1075717] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023] Open
Abstract
Trypanosoma cruzi, the causal agent of Chagas disease, has coexisted with humans for thousands of years. Therefore, the parasite has developed several mechanisms of antigenic variability that has allowed it to live inside the cells and evade the host immune response. Since T. cruzi displays an intracellular cycle-stage, our research team focused on providing insights into the CD8+ T cells immune response in chronic Chagas cardiomyopathy. We began our work in the 2000s studying parasite antigens that induce natural immune responses such as the KMP11 protein and TcTLE, its N-terminal derived peptide. Different approaches allowed us to reveal TcTLE peptide as a promiscuous CD8+ T cell epitope, able of inducing multifunctional cellular immune responses and eliciting a humoral response capable of decreasing parasite movement and infective capacity. Next, we demonstrated that as the disease progresses, total CD8+ T cells display a dysfunctional state characterized by a prolonged hyper-activation state along with an increase of inhibitory receptors (2B4, CD160, PD-1, TIM-3, CTLA-4) expression, an increase of specific terminal effector T cells (TTE), a decrease of proliferative capacity, a decrease of stem cell memory (TSCM) frequency, and a decrease of CD28 and CD3ζ expression. Thus, parasite-specific CD8+ T cells undergo clonal exhaustion, distinguished by an increase in late-differentiated cells, a mono-functional response, and enhanced expression of inhibitory receptors. Finally, it was found that anti-parasitic treatment induces an improved CD8+ T cell response in asymptomatic individuals, and a mouse animal model led us to establish a correlation between the quality of the CD8+ T cell responses and the outcome of chronic infection. In the future, using OMICs strategies, the identification of the specific cellular signals involved in disease progression will provide an invaluable resource for discovering new biomarkers of progression or new vaccine and immunotherapy strategies. Also, the inclusion of the TcTLE peptide in the rational design of epitope-based vaccines, the development of immunotherapy strategies using TSCM or the blocking of inhibitory receptors, and the use of the CD8+ T cell response quality to follow treatments, immunotherapies or vaccines, all are alternatives than could be explored in the fight against Chagas disease.
Collapse
Affiliation(s)
- Concepción J. Puerta
- Laboratory of Molecular Parasitology, Infectious Diseases Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Cuellar
- Clinical Laboratory Sciences Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Paola Lasso
- Laboratory of Molecular Parasitology, Infectious Diseases Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jose Mateus
- Laboratory of Molecular Parasitology, Infectious Diseases Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - John M. Gonzalez
- Group of Biomedical Sciences, School of Medicine, Universidad de Los Andes, Bogotá, Colombia
| |
Collapse
|
13
|
Kian M, Mirzavand S, Sharifzadeh S, Kalantari T, Ashrafmansouri M, Nasri F. Efficacy of Mesenchymal Stem Cells Therapy in Parasitic Infections: Are Anti-parasitic Drugs Combined with MSCs More Effective? Acta Parasitol 2022; 67:1487-1499. [DOI: 10.1007/s11686-022-00620-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 09/20/2022] [Indexed: 11/01/2022]
|
14
|
Scariot DB, Staneviciute A, Zhu J, Li X, Scott EA, Engman DM. Leishmaniasis and Chagas disease: Is there hope in nanotechnology to fight neglected tropical diseases? Front Cell Infect Microbiol 2022; 12:1000972. [PMID: 36189341 PMCID: PMC9523166 DOI: 10.3389/fcimb.2022.1000972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
Nanotechnology is revolutionizing many sectors of science, from food preservation to healthcare to energy applications. Since 1995, when the first nanomedicines started being commercialized, drug developers have relied on nanotechnology to improve the pharmacokinetic properties of bioactive molecules. The development of advanced nanomaterials has greatly enhanced drug discovery through improved pharmacotherapeutic effects and reduction of toxicity and side effects. Therefore, highly toxic treatments such as cancer chemotherapy, have benefited from nanotechnology. Considering the toxicity of the few therapeutic options to treat neglected tropical diseases, such as leishmaniasis and Chagas disease, nanotechnology has also been explored as a potential innovation to treat these diseases. However, despite the significant research progress over the years, the benefits of nanotechnology for both diseases are still limited to preliminary animal studies, raising the question about the clinical utility of nanomedicines in this field. From this perspective, this review aims to discuss recent nanotechnological developments, the advantages of nanoformulations over current leishmanicidal and trypanocidal drugs, limitations of nano-based drugs, and research gaps that still must be filled to make these novel drug delivery systems a reality for leishmaniasis and Chagas disease treatment.
Collapse
Affiliation(s)
- Debora B. Scariot
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Austeja Staneviciute
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Jennifer Zhu
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - Xiaomo Li
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, Northwestern University, Chicago, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, and Simpson Querrey Institute, Northwestern University, Evanston and Chicago, IL, United States
| | - David M. Engman
- Department of Pathology, Northwestern University, Chicago, IL, United States
| |
Collapse
|
15
|
Metabolomic Profiling of End-Stage Heart Failure Secondary to Chronic Chagas Cardiomyopathy. Int J Mol Sci 2022; 23:ijms231810456. [PMID: 36142367 PMCID: PMC9499603 DOI: 10.3390/ijms231810456] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic Chagas cardiomyopathy (CCC) is the most frequent and severe clinical form of chronic Chagas disease, representing one of the leading causes of morbidity and mortality in Latin America, and a growing global public health problem. There is currently no approved treatment for CCC; however, omics technologies have enabled significant progress to be made in the search for new therapeutic targets. The metabolic alterations associated with pathogenic mechanisms of CCC and their relationship to cellular and immunopathogenic processes in cardiac tissue remain largely unknown. This exploratory study aimed to evaluate the potential underlying pathogenic mechanisms in the failing myocardium of patients with end-stage heart failure (ESHF) secondary to CCC by applying an untargeted metabolomic profiling approach. Cardiac tissue samples from the left ventricle of patients with ESHF of CCC etiology (n = 7) and healthy donors (n = 7) were analyzed using liquid chromatography-mass spectrometry. Metabolite profiles showed altered branched-chain amino acid and acylcarnitine levels, decreased fatty acid uptake and oxidation, increased activity of the pentose phosphate pathway, dysregulation of the TCA cycle, and alterations in critical cellular antioxidant systems. These findings suggest processes of energy deficit, alterations in substrate availability, and enhanced production of reactive oxygen species in the affected myocardium. This profile potentially contributes to the development and maintenance of a chronic inflammatory state that leads to progression and severity of CCC. Further studies involving larger sample sizes and comparisons with heart failure patients without CCC are needed to validate these results, opening an avenue to investigate new therapeutic approaches for the treatment and prevention of progression of this unique and severe cardiomyopathy.
Collapse
|
16
|
Chaves AT, Oliveira ALGD, Guimarães NS, Magalhães IC, Menezes CADS, Rocha MODC. Galectin-3 and fibrosis intensity in Chronic Chagas Cardiomyopathy: a systematic review. Rev Inst Med Trop Sao Paulo 2022; 64:e45. [PMID: 35749417 PMCID: PMC9236203 DOI: 10.1590/s1678-9946202264045] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/15/2022] [Indexed: 11/24/2022] Open
Abstract
Chronic Chagas Cardiomyopathy (CCC) is the most prevalent type of myocarditis and the main clinical form of the Chagas disease, which has peculiarities such as focal inflammation, structural derangement, hypertrophy, dilation, and intense reparative fibrosis. Many cellular compounds contribute to CCC development. Galectin-3 is a partaker in inflammation and contributes to myocardial fibrosis formation. Some studies showed the connection between Galectin-3 and fibrosis in Chagas disease but are still inconclusive on the guidance for the early implementation of pharmacological therapy. This systematic review evaluated Galectin-3 as a biomarker for fibrosis intensity in CCC. Two independent reviewers have searched five databases (PubMed, EMBASE, Cochrane Library, Scopus, and Lilacs), using the following search terms: galectin-3, biomarkers, fibrosis, Chagas cardiomyopathy, and Chagas disease. Overall, seven studies met the inclusion criteria and made up this review. There were four trials conducted through animal model experiments and three trials with humans. Experimental data in mice indicate an association between Galectin-3 expression and fibrosis in CCC (75% of studies). Data from human studies showed no direct connection between myocardial fibrosis and Galectin-3 expression (80% of studies). Thus, human findings do not provide significant evidence indicating that Galectin-3 is related to fibrosis formation in Chagas disease. Based on the analyzed studies, it is suggested that Galectin-3 might not be a good fibrosis marker in CCC.
Collapse
Affiliation(s)
- Ana Thereza Chaves
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde: Medicina Tropical e Doenças Infecciosas, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Laura Grossi de Oliveira
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde: Medicina Tropical e Doenças Infecciosas, Belo Horizonte, Minas Gerais, Brazil
| | - Nathalia Sernizon Guimarães
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde: Medicina Tropical e Doenças Infecciosas, Belo Horizonte, Minas Gerais, Brazil
| | - Isabela Cristina Magalhães
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde: Medicina Tropical e Doenças Infecciosas, Belo Horizonte, Minas Gerais, Brazil
| | - Cristiane Alves da Silva Menezes
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, Minas Gerais, Brazil
| | - Manoel Otávio da Costa Rocha
- Universidade Federal de Minas Gerais, Faculdade de Medicina, Programa de Pós-Graduação em Ciências da Saúde: Medicina Tropical e Doenças Infecciosas, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
17
|
Pieralisi AV, Cevey ÁC, Penas FN, Prado N, Mori A, Gili M, Mirkin GA, Gagliardi J, Goren NB. Fenofibrate Increases the Population of Non-Classical Monocytes in Asymptomatic Chagas Disease Patients and Modulates Inflammatory Cytokines in PBMC. Front Cell Infect Microbiol 2022; 11:785166. [PMID: 35360222 PMCID: PMC8963737 DOI: 10.3389/fcimb.2021.785166] [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: 09/28/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic Chagas disease cardiomyopathy (CCC) is the most important clinical manifestation of infection with Trypanosma cruzi (T. cruzi) due to its frequency and effects on morbidity and mortality. Peripheral blood mononuclear cells (PBMC) infiltrate the tissue and differentiate into inflammatory macrophages. Advances in pathophysiology show that myeloid cell subpopulations contribute to cardiac homeostasis, emerging as possible therapeutic targets. We previously demonstrated that fenofibrate, PPARα agonist, controls inflammation, prevents fibrosis and improves cardiac function in a murine infection model. In this work we investigated the spontaneous release of inflammatory cytokines and chemokines, changes in the frequencies of monocyte subsets, and fenofibrate effects on PBMC of seropositive patients with different clinical stages of Chagas disease. The results show that PBMC from Chagas disease patients display higher levels of IL-12, TGF-β, IL-6, MCP1, and CCR2 than cells from uninfected individuals (HI), irrespectively of the clinical stage, asymptomatic (Asy) or with Chagas heart disease (CHD). Fenofibrate reduces the levels of pro-inflammatory mediators and CCR2 in both Asy and CHD patients. We found that CHD patients display a significantly higher percentage of classical monocytes in comparison with Asy patients and HI. Besides, Asy patients have a significantly higher percentage of non-classical monocytes than CHD patients or HI. However, no difference in the intermediate monocyte subpopulation was found between groups. Moreover, monocytes from Asy or CHD patients exhibit different responses upon stimulation in vitro with T. cruzi lysates and fenofibrate treatment. Stimulation with T. cruzi significantly increases the percentage of classical monocytes in the Asy group whereas the percentage of intermediate monocytes decreases. Besides, there are no changes in their frequencies in CHD or HI. Notably, stimulation with T. cruzi did not modify the frequency of the non-classical monocytes subpopulation in any of the groups studied. Moreover, fenofibrate treatment of T. cruzi-stimulated cells, increased the frequency of the non-classical subpopulation in Asy patients. Interestingly, fenofibrate restores CCR2 levels but does not modify HLA-DR expression in any groups. In conclusion, our results emphasize a potential role for fenofibrate as a modulator of monocyte subpopulations towards an anti-inflammatory and healing profile in different stages of chronic Chagas disease.
Collapse
Affiliation(s)
- Azul V. Pieralisi
- Universidad de Buenos Aires. Facultad de Medicina. Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
- CONICET Universidad de Buenos Aires. Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Ágata C. Cevey
- Universidad de Buenos Aires. Facultad de Medicina. Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
- CONICET Universidad de Buenos Aires. Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Federico N. Penas
- Universidad de Buenos Aires. Facultad de Medicina. Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
- CONICET Universidad de Buenos Aires. Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Nilda Prado
- Division of Cardiology, Hospital del Gobierno de la Ciudad de Buenos Aires "Dr. Cosme Argerich", Buenos Aires, Argentina
| | - Ana Mori
- Division of Cardiology, Hospital del Gobierno de la Ciudad de Buenos Aires "Dr. Cosme Argerich", Buenos Aires, Argentina
| | - Mónica Gili
- Hospital Municipal de Rehabilitación Respiratoria María Ferrer, Buenos Aires, Argentina
| | - Gerardo A. Mirkin
- Universidad de Buenos Aires. Facultad de Medicina. Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
- CONICET Universidad de Buenos Aires. Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Buenos Aires, Argentina
| | - Juan Gagliardi
- Division of Cardiology, Hospital del Gobierno de la Ciudad de Buenos Aires "Dr. Cosme Argerich", Buenos Aires, Argentina
| | - Nora B. Goren
- Universidad de Buenos Aires. Facultad de Medicina. Departamento de Microbiología, Parasitología e Inmunología, Buenos Aires, Argentina
- CONICET Universidad de Buenos Aires. Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
- *Correspondence: Nora B. Goren,
| |
Collapse
|
18
|
Experimental Combination Therapy with Amiodarone and Low-Dose Benznidazole in a Mouse Model of Trypanosoma cruzi Acute Infection. Microbiol Spectr 2022; 10:e0185221. [PMID: 35138142 PMCID: PMC8826820 DOI: 10.1128/spectrum.01852-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Chagas disease (CD), caused by Trypanosoma cruzi, affects approximately 6 to 7 million people in Latin America, with cardiomyopathy being the clinical manifestation most commonly associated with patient death during the acute phase. The etiological treatment of CD is restricted to benznidazole (Bz) and nifurtimox (Nif), which involve long periods of administration, frequent side effects, and low efficacy in the chronic phase. Thus, combined therapies emerge as an important tool in the treatment of CD, allowing the reduction of Bz dose and treatment duration. In this sense, amiodarone (AMD), the most efficient antiarrhythmic drug currently available and prescribed to CD patients, is a potential candidate for combined treatment due to its known trypanocidal activity. However, the efficacy of AMD during the acute phase of CD and its interaction with Bz or Nif are still unknown. In the present study, using a well-established murine model of the acute phase of CD, we observed that the Bz/AMD combination was more effective in reducing the peak parasitemia than both monotherapy treatments. Additionally, the Bz/AMD combination reduced (i) interleukin-6 (IL-6) levels in cardiac tissue, (ii) P-wave duration, and (iii) frequency of arrhythmia in infected animals and (iv) restored gap junction integrity in cardiac tissue. Therefore, our study validates AMD as a promising candidate for combined therapy with Bz, reinforcing the strategy of combined therapy for CD. IMPORTANCE Chagas disease affects approximately 6 to 7 million people worldwide, with cardiomyopathy being the clinical manifestation that most commonly leads to patient death. The etiological treatment of Chagas disease is limited to drugs (benznidazole and nifurtimox) with relatively high toxicity and therapeutic failures. In this sense, amiodarone, the most effective currently available antiarrhythmic drug prescribed to patients with Chagas disease, is a potential candidate for combined treatment due to its known trypanocidal effect. In the present study, we show that combined treatment with benznidazole and amiodarone improves the trypanocidal effect and reduces cardiac damage in acutely T. cruzi-infected mice.
Collapse
|
19
|
Hoffman KA, Villar MJ, Poveda C, Bottazzi ME, Hotez PJ, Tweardy DJ, Jones KM. Signal Transducer and Activator of Transcription-3 Modulation of Cardiac Pathology in Chronic Chagasic Cardiomyopathy. Front Cell Infect Microbiol 2021; 11:708325. [PMID: 34504808 PMCID: PMC8421853 DOI: 10.3389/fcimb.2021.708325] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/23/2021] [Indexed: 01/31/2023] Open
Abstract
Chronic Chagasic cardiomyopathy (CCC) is a severe clinical manifestation that develops in 30%–40% of individuals chronically infected with the protozoal parasite Trypanosoma cruzi and is thus an important public health problem. Parasite persistence during chronic infection drives pathologic changes in the heart, including myocardial inflammation and progressive fibrosis, that contribute to clinical disease. Clinical manifestations of CCC span a range of symptoms, including cardiac arrhythmias, thromboembolic disease, dilated cardiomyopathy, and heart failure. This study aimed to investigate the role of signal transducer and activator of transcription-3 (STAT3) in cardiac pathology in a mouse model of CCC. STAT3 is a known cellular mediator of collagen deposition and fibrosis. Mice were infected with T. cruzi and then treated daily from 70 to 91 days post infection (DPI) with TTI-101, a small molecule inhibitor of STAT3; benznidazole; a combination of benznidazole and TTI-101; or vehicle alone. Cardiac function was evaluated at the beginning and end of treatment by echocardiography. By the end of treatment, STAT3 inhibition with TTI-101 eliminated cardiac fibrosis and fibrosis biomarkers but increased cardiac inflammation; serum levels of interleukin-6 (IL-6), and IFN−γ; cardiac gene expression of STAT1 and nuclear factor-κB (NF-κB); and upregulation of IL-6 and Type I and Type II IFN responses. Concurrently, decreased heart function was measured by echocardiography and myocardial strain. These results indicate that STAT3 plays a critical role in the cardiac inflammatory–fibrotic axis during CCC.
Collapse
Affiliation(s)
- Kristyn A Hoffman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Maria Jose Villar
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States
| | - Cristina Poveda
- Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States
| | - Maria Elena Bottazzi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States.,Department of Biology, Baylor University, Waco, TX, United States
| | - Peter J Hotez
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States.,Department of Biology, Baylor University, Waco, TX, United States
| | - David J Tweardy
- Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine and Department of Molecular & Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kathryn M Jones
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States.,Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, TX, United States.,Texas Children's Hospital Center for Vaccine Development, Houston, TX, United States
| |
Collapse
|
20
|
Apt W, Llancaqueo M, Zulantay I, Canals M, Kara S, Arribada A, Muñoz G, Martínez G. Clinical, electrocardiographic and echocardiographic evolution of chronic Chagas disease treated with nifurtimox on prolonged follow-up in Chile: observational study. J Glob Antimicrob Resist 2021; 27:160-166. [PMID: 34496323 DOI: 10.1016/j.jgar.2021.08.012] [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: 01/24/2021] [Revised: 07/07/2021] [Accepted: 08/19/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES This study aimed to describe the electrocardiographic and echocardiographic status of chronic Chagas disease (cChD) patients treated with nifurtimox. METHODS An observational study was performed in 146 cChD patients followed over a mean of 7.9 years. RESULTS Of the 146 patients, 41 (28.1%) with normal electrocardiogram (ECG) at baseline maintained this condition, 34 (23.3%) with altered ECG at baseline normalised the alterations, and 46 (31.5%) with ECG abnormalities at baseline maintained this condition [23 (15.8%) with small alterations]. Finally, 25 cases (17.1%) in indeterminate phase altered the ECG. Differences before and after follow-up (P < 0.001) were found. The percentage of beneficial treatment was different than expected by chance (Z = 4.8; P < 0.001) and the annual percentage of cases that developed ECG alterations was lower than that of a historical cohort of untreated patients (P < 0.001). An echocardiogram was performed in 68 patients with baseline ECG alterations. The ejection fraction (EF) was normal in 57 (83.8%) and abnormal in 11 (16.2%). In 38 patients with ECG abnormalities that did not progress after treatment, EF and segmental motility (SM) were normal in 31 (81.6%) and 26 (68.4%), respectively. In 17 patients with ECG abnormalities, EF and SM were normal in 15 (88.2%) and 14 (82.4%) cases, respectively. CONCLUSION Less progression to cardiomyopathy compared with a historical untreated cohort as well as the EF/SM results in patients with abnormal ECG that did not progress and in indeterminate cChD that altered the ECG suggests a beneficial effect of nifurtimox.
Collapse
Affiliation(s)
- Werner Apt
- Laboratory of Basic-Clinical Parasitology, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Marcelo Llancaqueo
- Department of Cardiology, Clinic Hospital University of Chile, Santiago, Chile
| | - Inés Zulantay
- Laboratory of Basic-Clinical Parasitology, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Mauricio Canals
- School of Public Health, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sergio Kara
- Service of Cardiology, San José Hospital, Santiago, Chile
| | | | - Gabriela Muñoz
- Laboratory of Basic-Clinical Parasitology, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Gabriela Martínez
- Laboratory of Basic-Clinical Parasitology, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
21
|
das Dores Pereira R, Rabelo RAN, Leite PG, Cramer A, Botelho AFM, Cruz JS, Régis WCB, Perretti M, Teixeira MM, Machado FS. Role of formyl peptide receptor 2 (FPR2) in modulating immune response and heart inflammation in an experimental model of acute and chronic Chagas disease. Cell Immunol 2021; 369:104427. [PMID: 34482259 DOI: 10.1016/j.cellimm.2021.104427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/15/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022]
Abstract
Chagas disease is an important disease of the heart. Lipoxins have important regulatory functions in host immune response (IR). Herein, we examined whether the receptor for lipoxin A4, the formyl peptide receptor (FPR) 2, had an effect on Trypanosoma cruzi infection. In vitro, FPR2 deficiency or inhibition improved the activity of macrophages against T. cruzi. In vivo, during the acute phase, the absence of FPR2 reduced parasitemia and increased type 2 macrophages, type 2 neutrophils, and IL-10-producing dendritic cells. Moreover, the acquired IR was characterized by greater proportions of Th1/Th2/Treg, and IFNγ-producing CD8+T cells, and reductions in Th17 and IL-17-producing CD8+T cells. However, during the chronic phase, FPR2 deficient mice presented and increased inflammatory profile regarding innate and acquired IR cells (Th1/IFN-γ-producing CD8+T cells). Notably, FPR2 deficiency resulted in increased myocarditis and impaired heart function. Collectively, our data suggested that FPR2 is important for the orchestration of IR and prevention of severe T. cruzi-induced disease.
Collapse
Affiliation(s)
- Rafaela das Dores Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences, Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rayane Aparecida Nonato Rabelo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences, Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Paulo Gaio Leite
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Allysson Cramer
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Flávia Machado Botelho
- Departament of Veterinary Medicine, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiânia, Brazil
| | - Jader Santos Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Wiliam César Bento Régis
- Postgraduate Program in Vertebrate Biology at the Pontifical Catholic University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences, Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Program in Health Sciences, Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
22
|
Hoffman K, Liu Z, Hossain E, Bottazzi ME, Hotez PJ, Jones KM, McCall LI. Alterations to the Cardiac Metabolome Induced by Chronic T. cruzi Infection Relate to the Degree of Cardiac Pathology. ACS Infect Dis 2021; 7:1638-1649. [PMID: 33843195 PMCID: PMC8588157 DOI: 10.1021/acsinfecdis.0c00816] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic Chagasic cardiomyopathy (CCC) is a Neglected Tropical Disease caused by the parasite Trypanosoma cruzi. The pathognomonic findings in symptomatic CCC patients and animal models includes diffuse cardiac fibrosis and inflammation with persistent parasite presence in the heart. This study investigated chemical alterations in different regions of the heart in relation to cardiac pathology indicators to better understand the long-term pathogenesis of this neglected disease. We used data from echocardiography, fibrosis biomarkers, and histopathological analysis to fully evaluate cardiac pathology. Metabolites isolated from the pericardial and endocardial sides of the right ventricular myocardium were analyzed by liquid chromatography tandem mass spectrometry. The endocardial sections contained significantly less cardiac inflammation and fibrosis than the pericardial sections. Cardiac levels of acylcarnitines, phosphocholines, and other metabolites were significantly disrupted in accordance with cardiac fibrosis, inflammation, and serum fibrosis biomarker levels. These findings have potential implications in treatment and monitoring for CCC patients.
Collapse
Affiliation(s)
- Kristyn Hoffman
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Zongyuan Liu
- Department of Chemistry and Biochemistry and Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Ekram Hossain
- Department of Chemistry and Biochemistry and Laboratories of Molecular Anthropology and Microbiome Research, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Maria Elena Bottazzi
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States; Texas Children’s Hospital Center for Vaccine Development, Houston, Texas 77030, United States; Department of Biology, Baylor University, Waco, Texas 76798, United States
| | - Peter J. Hotez
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States; Texas Children’s Hospital Center for Vaccine Development, Houston, Texas 77030, United States; Department of Biology, Baylor University, Waco, Texas 76798, United States
| | - Kathryn M. Jones
- Department of Molecular Virology and Microbiology and Department of Pediatrics, Section of Tropical Medicine, Baylor College of Medicine, Houston, Texas 77030, United States; Texas Children’s Hospital Center for Vaccine Development, Houston, Texas 77030, United States
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, Laboratories of Molecular Anthropology and Microbiome Research, and Department of Microbiology and Plant Biology, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
23
|
de Souza DMS, Silva MC, Farias SEB, Menezes APDJ, Milanezi CM, Lúcio KDP, Paiva NCN, de Abreu PM, Costa DC, Pinto KMDC, Costa GDP, Silva JS, Talvani A. Diet Rich in Lard Promotes a Metabolic Environment Favorable to Trypanosoma cruzi Growth. Front Cardiovasc Med 2021; 8:667580. [PMID: 34113663 PMCID: PMC8185140 DOI: 10.3389/fcvm.2021.667580] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/15/2021] [Indexed: 01/24/2023] Open
Abstract
Background: Trypanosoma cruzi is a protozoan parasite that causes Chagas disease and affects 6-7 million people mainly in Latin America and worldwide. Here, we investigated the effects of hyperlipidic diets, mainly composed of olive oil or lard on experimental T. cruzi infection. C57BL/6 mice were fed two different dietary types in which the main sources of fatty acids were either monounsaturated (olive oil diet) or saturated (lard diet). Methods: After 60 days on the diet, mice were infected with 50 trypomastigote forms of T. cruzi Colombian strain. We evaluated the systemic and tissue parasitism, tissue inflammation, and the redox status of mice after 30 days of infection. Results: Lipid levels in the liver of mice fed with the lard diet increased compared with that of the mice fed with olive oil or normolipidic diets. The lard diet group presented with an increased parasitic load in the heart and adipose tissues following infection as well as an increased expression of Tlr2 and Tlr9 in the heart. However, no changes were seen in the survival rates across the dietary groups. Infected mice receiving all diets presented comparable levels of recruited inflammatory cells at 30 days post-infection but, at this time, we observed lard diet inducing an overproduction of CCL2 in the cardiac tissue and its inhibition in the adipose tissue. T. cruzi infection altered liver antioxidant levels in mice, with the lard diet group demonstrating decreased catalase (CAT) activity compared with that of other dietary groups. Conclusions: Our data demonstrated that T. cruzi growth is more favorable on tissue of mice subjected to the lard diet. Our findings supported our hypothesis of a relationship between the source of dietary lipids and parasite-induced immunopathology.
Collapse
Affiliation(s)
- Débora Maria Soares de Souza
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Biological Science Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Maria Cláudia Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Silvia Elvira Barros Farias
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Ana Paula de J Menezes
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Cristiane Maria Milanezi
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Karine de P Lúcio
- Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Nívia Carolina N Paiva
- Center of Research in Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Paula Melo de Abreu
- Biological Science Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Daniela Caldeira Costa
- Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil.,Laboratory of Metabolic Biochemistry, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Kelerson Mauro de Castro Pinto
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,School of Physical Education, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - João Santana Silva
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil.,Fiocruz-Bi-Institutional Translational Medicine Plataform, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation, Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health and Nutrition Post-graduate Program, Federal University of Ouro Preto, Ouro Preto, Brazil.,Health Science, Infectology and Tropical Medicine Post-graduate Program, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
24
|
Rial MS, Seremeta KP, Esteva MI, Búa J, Salomon CJ, Fichera LE. In vitro studies and preclinical evaluation of benznidazole microparticles in the acute Trypanosoma cruzi murine model. Parasitology 2021; 148:566-575. [PMID: 33298212 PMCID: PMC10950374 DOI: 10.1017/s0031182020002310] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 01/28/2023]
Abstract
Chagas disease is a serious parasitic infection caused by Trypanosoma cruzi. Unfortunately, the current chemotherapeutic tools are not enough to combat the infection. The aim of this study was to evaluate the trypanocidal activity of benznidazole-loaded microparticles during the acute phase of Chagas infection in an experimental murine model. Microparticles were prepared by spray-drying using copolymers derived from esters of acrylic and methacrylic acids as carriers. Dissolution efficiency of the formulations was up to 3.80-fold greater than that of raw benznidazole. Stability assay showed no significant difference (P > 0.05) in the loading capacity of microparticles for 3 years. Cell cultures showed no visible morphological changes or destabilization of the cell membrane nor haemolysis was observed in defibrinated human blood after microparticles treatment. Mice with acute lethal infection survived 100% after 30 days of treatment with benznidazole microparticles (50 mg kg-1 day-1). Furthermore, no detectable parasite load measured by quantitative polymerase chain reaction and lower levels of T. cruzi-specific antibodies by enzyme-linked immunosorbent assay were found in those mice. A significant decrease in the inflammation of heart tissue after treatment with these microparticles was observed, in comparison with the inflammatory damage observed in both infected mice treated with raw benznidazole and untreated infected mice. Therefore, these polymeric formulations are an attractive approach to treat Chagas disease.
Collapse
Affiliation(s)
- Marcela S. Rial
- Instituto Nacional de Parasitología Dr M. Fatala Chaben, ANLIS CG Malbrán, Ministerio de Salud, Av. Paseo Colón 568, Ciudad de Buenos Aires, Argentina
| | - Katia P. Seremeta
- Departamento de Ciencias Básicas y Aplicadas, Universidad Nacional del Chaco Austral, Cte. Fernández 755, 3700, Pcia. Roque Sáenz Peña, Chaco, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mónica I. Esteva
- Instituto Nacional de Parasitología Dr M. Fatala Chaben, ANLIS CG Malbrán, Ministerio de Salud, Av. Paseo Colón 568, Ciudad de Buenos Aires, Argentina
| | - Jacqueline Búa
- Instituto Nacional de Parasitología Dr M. Fatala Chaben, ANLIS CG Malbrán, Ministerio de Salud, Av. Paseo Colón 568, Ciudad de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Claudio J. Salomon
- Instituto de Química Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (IQUIR-CONICET), Suipacha 531, 2000, Rosario, Argentina
- Área Técnica Farmacéutica, Departamento de Farmacia, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Laura E. Fichera
- Instituto Nacional de Parasitología Dr M. Fatala Chaben, ANLIS CG Malbrán, Ministerio de Salud, Av. Paseo Colón 568, Ciudad de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
25
|
Milduberger N, Bustos PL, González C, Perrone AE, Postan M, Bua J. Trypanosoma cruzi infection in Cyclophilin D deficient mice. Exp Parasitol 2021; 220:108044. [PMID: 33253715 DOI: 10.1016/j.exppara.2020.108044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/10/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022]
Abstract
Trypanosoma cruzi is the causative agent of Chagas disease, which is endemic in Latin America and around the world through mother to child transmission. The heart is the organ most frequently affected in the chronic stage of the human infection and depends on mitochondria for the required energy for its activity. Cyclophilins are involved in protein folding and the mitochondrial isoform, Cyclophilin D (CyPD), has a crucial role in the opening of the mitochondrial permeability transition pore. In the present study, we infected CyPD deficient mice, with ablation of the Ppif gene, with T. cruzi parasites and the course of the infection was analyzed. Parasite load, quantified by PCR, was significantly lower in skeletal and cardiac tissues of Ppif-/- mice compared to wild type mice. In vitro cultured cardiomyocytes and macrophages from mice lacking CyPD exhibited lower percentage of infected cells and number of intracellular parasites than those observed for wild type mice. Although histopathological analysis of heart and mRNA of heart cytokines showed differences between T. cruzi-infected mice compared to the uninfected animals, no significant differences were found mice due to the ablation of the Ppif gene. Our results suggest that cells deficient for mitochondrial CyPD, inhibited for the mitochondrial membrane potential collapse, reduces the severity of parasite aggression and spread of cellular infection.
Collapse
MESH Headings
- Animals
- Chagas Disease/parasitology
- Peptidyl-Prolyl Isomerase F/deficiency
- Cytokines/analysis
- Cytokines/genetics
- DNA, Protozoan/isolation & purification
- Heart/parasitology
- Liver/pathology
- Macrophages, Peritoneal/cytology
- Macrophages, Peritoneal/parasitology
- Mice
- Mice, Inbred C57BL
- Muscle, Skeletal/parasitology
- Muscle, Skeletal/pathology
- Myocardium/pathology
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/parasitology
- Parasite Load
- RNA, Messenger/analysis
- RNA, Protozoan/analysis
- RNA, Protozoan/isolation & purification
- Spleen/pathology
- Trypanosoma cruzi/genetics
- Trypanosoma cruzi/physiology
Collapse
Affiliation(s)
- Natalia Milduberger
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Paseo Colón 568, PC 1063, Buenos Aires, Argentina
| | - Patricia L Bustos
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Paseo Colón 568, PC 1063, Buenos Aires, Argentina
| | - Carolina González
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Paseo Colón 568, PC 1063, Buenos Aires, Argentina; Centro de Altos Estudios en Ciencias Humanas y de La Salud (CAECIHS), Universidad Abierta Interamericana, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina
| | - Alina E Perrone
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Paseo Colón 568, PC 1063, Buenos Aires, Argentina
| | - Miriam Postan
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Paseo Colón 568, PC 1063, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina
| | - Jacqueline Bua
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" - ANLIS C. G. Malbrán, Paseo Colón 568, PC 1063, Buenos Aires, Argentina; Centro de Altos Estudios en Ciencias Humanas y de La Salud (CAECIHS), Universidad Abierta Interamericana, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas, Buenos Aires, Argentina.
| |
Collapse
|
26
|
Ledezma AP, Blandon R, Schijman AG, Benatar A, Saldaña A, Osuna A. Mixed infections by different Trypanosoma cruzi discrete typing units among Chagas disease patients in an endemic community in Panama. PLoS One 2020; 15:e0241921. [PMID: 33180799 PMCID: PMC7660484 DOI: 10.1371/journal.pone.0241921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/22/2020] [Indexed: 01/02/2023] Open
Abstract
Background Trypanosoma cruzi, the hemoparasite that causes Chagas disease, is divided into six Discrete Typing Units or DTUs: TcI-TcVI plus Tcbat. This genetic diversity is based on ecobiological and clinical characteristics associated with particular populations of the parasite. The main objective of this study was the identification of DTUs in patients with chronic chagasic infections from a mountainous rural community in the eastern region of Panama. Methods A total of 106 patients were tested for Chagas disease with three serological tests (ELISA, rapid test, and Western blot). Molecular diagnosis and DTU typing were carried out by conventional PCRs and qPCR targeting different genomic markers, respectively. As a control sample for the typing, 28 patients suspected to be chagasic from the metropolitan area of Panama City were included. Results Results showed a positivity in the evaluated patients of 42.3% (33/78); high compared to other endemic regions in the country. In the control group, 20/28 (71.43%) patients presented positive serology. The typing of samples from rural patients showed that 78.78% (26/33) corresponded to TcI, while 9.09% (3/33) were mixed infections (TcI plus TcII/V/VI). Seventy-five percent (15/20) of the patients in the control group presented TcI, and in five samples it was not possible to typify the T. cruzi genotype involved. Conclusions These results confirm that TcI is the main DTU of T. cruzi present in chronic chagasic patients from Panama. However, the circulation of other genotypes (TcII/V/VI) in this country is described for the first time. The eco-epidemiological characteristics that condition the circulation of TcII/V/VI, as well as the immune and clinical impact of mixed infections in this remote mountainous region should be investigated, which will help local action programs in the surveillance, prevention, and management of Chagas disease.
Collapse
Affiliation(s)
- Alexa Prescilla Ledezma
- Institute of Biotechnology, Department of Parasitology, University of Granada, Granada, Spain.,Center for Research and Diagnosis of Parasitic Diseases (CIDEP), Faculty of Medicine, University of Panama, Panama, Panama
| | | | - Alejandro G Schijman
- Laboratory of Molecular Biology of Chagas Disease, Institute of Research in Genetic Engineering and Molecular Biology "Dr Héctor Torres" (INGEBI-CONICET), Buenos Aires, Argentina
| | - Alejandro Benatar
- Laboratory of Molecular Biology of Chagas Disease, Institute of Research in Genetic Engineering and Molecular Biology "Dr Héctor Torres" (INGEBI-CONICET), Buenos Aires, Argentina
| | - Azael Saldaña
- Center for Research and Diagnosis of Parasitic Diseases (CIDEP), Faculty of Medicine, University of Panama, Panama, Panama.,Gorgas Memorial Institute of Health Studies (ICGES), Panama, Panama
| | - Antonio Osuna
- Institute of Biotechnology, Department of Parasitology, University of Granada, Granada, Spain
| |
Collapse
|
27
|
A novel Trypanosoma cruzi secreted antigen as a potential biomarker of Chagas disease. Sci Rep 2020; 10:19591. [PMID: 33177582 PMCID: PMC7658208 DOI: 10.1038/s41598-020-76508-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 10/26/2020] [Indexed: 01/30/2023] Open
Abstract
Chagas drug discovery has been hampered by a lack of validated assays to establish treatment efficacy in pre-clinical animal models and in patients infected with T. cruzi. Reduced levels of parasite secreted antigens in the blood of infected hosts could be used to demonstrate treatment efficacy. A published proteomic study of parasite secreted antigens identified the hypothetical protein Tc_5171 as a secreted antigen. In this report, we developed Tc_5171 specific antibodies and showed that the native protein was expressed by the three life cycle stages of the parasite. Anti-peptide antibodies were able to detect the parasite antigen in blood of infected mice during the acute and the chronic phase of infection. Benznidazole treatment of infected mice significantly reduced their blood antigen levels. Of clinical significance, patients diagnosed with Chagas disease, either asymptomatic or with cardiac clinical symptoms had significantly higher Tc_5171 antigen levels compared to endemic controls. Pair-wise analysis, before and after Benznidazole treatment, of patients with asymptomatic Chagas disease showed a significant reduction in antigen levels post treatment. Taken together, our results indicate that Tc_5171 could be used as a novel biomarker of Chagas disease for diagnosis and to assess treatment efficacy.
Collapse
|
28
|
Trypanosoma cruzi Induces the PARP1/AP-1 Pathway for Upregulation of Metalloproteinases and Transforming Growth Factor β in Macrophages: Role in Cardiac Fibroblast Differentiation and Fibrosis in Chagas Disease. mBio 2020; 11:mBio.01853-20. [PMID: 33172999 PMCID: PMC7667027 DOI: 10.1128/mbio.01853-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiomyopathy is the most important clinical manifestation of T. cruzi-driven CD. Recent studies have suggested the detrimental role of the matrix metalloproteinases MMP2 and MMP9 in extracellular matrix (ECM) degradation during cardiac remodeling in T. cruzi infection. Peripheral TGF-β levels are increased in clinically symptomatic CD patients over those in clinically asymptomatic seropositive individuals. We provide the first evidence that during T. cruzi infection, Mϕ release of MMP2 and MMP9 plays an active role in activation of TGF-β signaling of ECM remodeling and cardiac fibroblast-to-myofibroblast differentiation. We also determined that PARP1 signals c-Fos- and JunB-mediated AP-1 transcriptional activation of profibrotic gene expression and demonstrated the significance of PARP1 inhibition in controlling chronic fibrosis in Chagas disease. Our study provides a promising therapeutic approach for controlling T. cruzi-driven fibroblast differentiation in CD by PARP1 inhibitors through modulation of the Mϕ signaling of the AP-1–MMP9–TGF-β pathway. Chagas disease (CD), caused by Trypanosoma cruzi, is a degenerative heart condition. In the present study, we investigated the role of poly [ADP-ribose] polymerase 1/activator protein 1 (PARP1/AP-1) in upregulation of profibrotic macrophages (Mϕ) and subsequent development of cardiac fibrosis in CD. We used in vitro and in vivo models of T. cruzi infection and chemical and genetic inhibition of Parp1 to examine the molecular mechanisms by which Mϕ might augment profibrotic events in CD. Cultured (RAW 264.7 and THP-1) Mϕ infected with T. cruzi and primary cardiac and splenic Mϕ of chronically infected mice exhibited a significant increase in the expression, activity, and release of metalloproteinases (MMP2, MMP9, and MMP12) and the cytokine transforming growth factor β (TGF-β). Mϕ release of MMPs and TGF-β signaled the cardiac fibroblast to myofibroblast differentiation, as evidenced by a shift from S100A4 to alpha smooth muscle actin (α-SMA) expression. Incubation of infected Mϕ with MMP2 and MMP9 inhibitors resulted in 60 to 74% decline in TGF-β release, and MMP9 and PARP1 inhibitors resulted in 57 to 70% decline in Mϕ TGF-β-driven cardiac fibroblast differentiation. Likewise, histological studies showed a 12- to 16-fold increase in myocardial expression of CD68 (Mϕ marker) and its colocalization with MMP9/TGF-β, galectin-3, and vimentin in wild-type mice with CD. In comparison, chronically infected Parp1−/− mice exhibited a >50% decline in myocardial levels of Mϕ and associated fibrosis markers. Further study showed that PARP1 synergized with c-Fos and JunB AP-1 family members for transcriptional activation of profibrotic response after T. cruzi infection. We conclude that PARP1 inhibition offers a potential therapy for controlling the T. cruzi-driven fibroblast differentiation in CD through modulation of the Mϕ signaling of the AP-1–MMP9–TGF-β pathway.
Collapse
|
29
|
Abstract
As the global COVID-19 pandemic advances, it increasingly impacts those vulnerable populations who already bear a heavy burden of neglected tropical disease. Chagas disease (CD), a neglected parasitic infection, is of particular concern because of its potential to cause cardiac, gastrointestinal, and other complications which could increase susceptibility to COVID-19. The over one million people worldwide with chronic Chagas cardiomyopathy require special consideration because of COVID-19’s potential impact on the heart, yet the pandemic also affects treatment provision to people with acute or chronic indeterminate CD. In this document, a follow-up to the WHF-IASC Roadmap on CD, we assess the implications of coinfection with SARS-CoV-2 and Trypanosoma cruzi, the etiological agent of CD. Based on the limited evidence available, we provide preliminary guidance for testing, treatment, and management of patients affected by both diseases, while highlighting emerging healthcare access challenges and future research needs.
Collapse
|
30
|
Chagas cardiomyopathy and heart failure: From epidemiology to treatment. Rev Port Cardiol 2020; 39:279-289. [PMID: 32532535 DOI: 10.1016/j.repc.2019.12.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 11/10/2019] [Accepted: 12/01/2019] [Indexed: 12/28/2022] Open
Abstract
Chagas disease is among the neglected tropical diseases recognized by the World Health Organization that have received insufficient attention from governments and health agencies. Chagas disease is endemic in 21 Latin America regions. Due to globalization and increased migration, it has crossed borders and reached other regions including North America and Europe. The clinical presentation of the disease is highly variable, from general symptoms to severe cardiac involvement that can culminate in heart failure. Chagas heart disease is multifactorial, and can include dilated cardiomyopathy, thromboembolic phenomena, and arrhythmias that may lead to sudden death. Diagnosis is by methods such as enzyme-linked immunosorbent assay (ELISA) and the degree of cardiac involvement should be investigated with complementary exams including ECG, chest radiography and electrophysiological study. There have been insufficient studies on which to base specific treatment for heart failure due to Chagas disease. Treatment should therefore be derived from guidelines for heart failure that are not specific for this disease. Heart transplantation is a viable option with satisfactory success rates that has improved survival.
Collapse
|
31
|
Santos É, Menezes Falcão L. Chagas cardiomyopathy and heart failure: From epidemiology to treatment. REVISTA PORTUGUESA DE CARDIOLOGIA (ENGLISH EDITION) 2020. [DOI: 10.1016/j.repce.2020.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
32
|
Echeverría LE, Marcus R, Novick G, Sosa-Estani S, Ralston K, Zaidel EJ, Forsyth C, RIbeiro ALP, Mendoza I, Falconi ML, Mitelman J, Morillo CA, Pereiro AC, Pinazo MJ, Salvatella R, Martinez F, Perel P, Liprandi ÁS, Piñeiro DJ, Molina GR. WHF IASC Roadmap on Chagas Disease. Glob Heart 2020; 15:26. [PMID: 32489799 PMCID: PMC7218776 DOI: 10.5334/gh.484] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/17/2020] [Indexed: 02/06/2023] Open
Abstract
Background Chagas Disease is a neglected tropical disease caused by the protozoan Trypanosoma cruzi, with some of the most serious manifestations affecting the cardiovascular system. It is a chronic, stigmatizing condition, closely associated with poverty and affecting close to 6 million people globally. Although historically the disease was limited to endemic areas of Latin America recent years have seen an increasing global spread. In addition to the morbidity and mortality associated with the disease, the social and economic burdens on individuals and society are substantial. Often called the 'silent killer', Chagas disease is characterized by a long, asymptomatic phase in affected individuals. Approximately 30% then go on develop chronic Chagas cardiomyopathy and other serious cardiac complications such as stroke, rhythm disturbances and severe heart failure. Methods In a collaboration of the World Hearth Federation (WHF) and the Inter-American Society of Cardiology (IASC) a writing group consisting of 20 diverse experts on Chagas disease (CD) was convened. The group provided up to date expert knowledge based on their area of expertise. An extensive review of the literature describing obstacles to diagnosis and treatment of CD along with proposed solutions was conducted. A survey was sent to all WHF Members and, using snowball sampling to widen the consultation, to a variety of health care professionals working in the CD global health community. The results were analyzed, open comments were reviewed and consolidated, and the findings were incorporated into this document, thus ensuring a consensus representation. Results The WHF IASC Roadmap on Chagas Disease offers a comprehensive summary of current knowledge on prevention, diagnosis and management of the disease. In providing an analysis of 'roadblocks' in access to comprehensive care for Chagas disease patients, the document serves as a framework from which strategies for implementation such as national plans can be formulated. Several dimensions are considered in the analysis: healthcare system capabilities, governance, financing, community awareness and advocacy. Conclusion The WHF IASC Roadmap proposes strategies and evidence-based solutions for healthcare professionals, health authorities and governments to help overcome the barriers to comprehensive care for Chagas disease patients. This roadmap describes an ideal patient care pathway, and explores the roadblocks along the way, offering potential solutions based on available research and examples in practice. It represents a call to action to decision-makers and health care professionals to step up efforts to eradicate Chagas disease.
Collapse
Affiliation(s)
| | - Rachel Marcus
- LASOCHA, Washington DC, US
- Medstar Union Memorial Hospital, Baltimore, MD, US
| | - Gabriel Novick
- Swiss Medical Group, Buenos Aires, AR
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, US
| | - Sergio Sosa-Estani
- Drugs for Neglected Diseases initiative-Latin America, Rio de Janeiro, BR
| | | | - Ezequiel Jose Zaidel
- Sanatorio Güemes, Buenos Aires, AR
- Pharmacology Department, School of Medicine, University of Buenos Aires, Buenos Aires, AR
| | - Colin Forsyth
- Drugs for Neglected Diseases initiative-Latin America, Rio de Janeiro, BR
| | - Antonio Luiz P. RIbeiro
- Internal Medicine Department, School of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, BR
- Hospital das Clínicas, UFMG, Belo Horizonte, BR
| | | | - Mariano Luis Falconi
- Cardiology Division, Italian Hospital of Buenos Aires, Buenos Aires, AR
- University Institute of the Italian Hospital of Buenos Aires, Buenos Aires, AR
| | - Jorge Mitelman
- Faculty of Medicine, University of Buenos Aires, Buenos Aires, AR
- School of Medicine, Barcélo University, Buenos Aires, AR
| | - Carlos A. Morillo
- Department of Cardiac Sciences, Cumming School of Medicine Division of Cardiology, Libin Cardiovascular Institute, University of Calgary, Calgary, CA
- Southeastern Alberta Region, Alberta Health Services, Foothills Medical Centre, CA
| | | | | | | | - Felipe Martinez
- National University of Cordoba, Cordoba, AR
- DAMIC Institute/Rusculleda Foundation, Cordoba, AR
| | - Pablo Perel
- World Heart Federation, Geneva, CH
- Centre for Global Chronic Conditions, London School of Hygiene and Tropical Medicine, London, GB
| | - Álvaro Sosa Liprandi
- Sanatorio Güemes, Buenos Aires, AR
- Medical School of Cardiology, University of Buenos Aires, Buenos Aires, AR
| | | | | |
Collapse
|
33
|
Ayyappan JP, Lizardo K, Wang S, Yurkow E, Nagajyothi JF. Inhibition of SREBP Improves Cardiac Lipidopathy, Improves Endoplasmic Reticulum Stress, and Modulates Chronic Chagas Cardiomyopathy. J Am Heart Assoc 2020; 9:e014255. [PMID: 31973605 PMCID: PMC7033903 DOI: 10.1161/jaha.119.014255] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Trypanosoma cruzi is an intracellular parasite that causes debilitating chronic Chagas cardiomyopathy (CCM), for which there is no effective drug or vaccine. Previously, we demonstrated increased cardiac lipid accumulation and endoplasmic reticulum stress in mice with CCM. Increased endoplasmic reticulum stress may lead to uncontrolled SREBP (sterol regulatory element-binding protein) activation and lipotoxicity in the myocardium during the intermediate stage of infection and result in progression to chronic CCM. Therefore, we investigated whether inhibiting SREBP activation modulates CCM progression in T cruzi-infected mice. Methods and Results T cruzi-infected cultured cardiomyocytes (3:1 multiplicity of infection; 24 hours postinfection) were incubated with betulin (3 μmol/L per mL), an SREBP inhibitor, for 24 hours. Quantitative polymerase chain reaction and Western blotting analyses demonstrated a significant reduction in SREBP activation, lipid biosynthesis, and endoplasmic reticulum stress in betulin-treated infected cells compared with untreated cells. T cruzi infected (103 trypomastigotes of the Brazil strain) Swiss mice were fed a customized diet containing betulin during the intermediate stage (40 days postinfection) until the chronic stage (120 DPI). Cardiac ultrasound imaging and histological and biochemical analyses demonstrated anatomical and functional improvements in betulin-treated, infected mice compared with untreated controls: we observed a significant reduction in cholesterol/fatty acid synthesis that may result in the observed cardiac reduction in cardiac lipid accumulation, mitochondrial and endoplasmic reticulum stress, and ventricular enlargement. Conclusions Our study (in vitro and vivo) demonstrates that inhibition of cardiac SREBP activation reduces cardiac damage during T cruzi infection and modulates CCM in a murine Chagas model.
Collapse
Affiliation(s)
- Janeesh Plakkal Ayyappan
- Department of Microbiology, Biochemistry and Molecular Genetics Public Health Research Institute New Jersey Medical School Newark NY
| | - Kezia Lizardo
- Department of Microbiology, Biochemistry and Molecular Genetics Public Health Research Institute New Jersey Medical School Newark NY
| | - Sean Wang
- Rutgers Molecular Imaging Center Piscataway NJ
| | | | - Jyothi F Nagajyothi
- Department of Microbiology, Biochemistry and Molecular Genetics Public Health Research Institute New Jersey Medical School Newark NY
| |
Collapse
|
34
|
Duque TLA, Cascabulho CM, Oliveira GM, Henriques-Pons A, Menna-Barreto RFS. Rapamycin Treatment Reduces Acute Myocarditis Induced by Trypanosoma cruzi Infection. J Innate Immun 2019; 12:321-332. [PMID: 31801138 DOI: 10.1159/000504322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
Chagas disease affects millions of people mainly in Latin America and is a protozoan illness caused by the parasite Trypanosoma cruzi. Chagasic cardiomyopathy is the leading cause of mortality of infected patients, due to compromised electrical and mechanical cardiac function induced by tissue remodeling, especially fibrosis, and lymphocytic infiltration. Some cellular biochemical pathways can be protective to the heart, and we tested if the in vivo activation of the autophagic machinery by rapamycin could reduce parasite-induced myocarditis. Regarding the expression of LC3, an autophagy marker, we observed its upregulation in the cardiac tissue of infected untreated mice. However, after rapamycin treatment, an autophagy inducer, infected mice showed reduced electrical cardiac dysfunctions, myocarditis, cardiac damage, and reduced production of pro-inflammatory cytokines by the heart. On the other hand, the parasite's life cycle was not affected, and we observed no modulations in cardiac tissue or blood parasitemia. Our data indicate that, at least partially, autophagy induction controls inflammation in the heart¸ illustrating the complexity of the pathways that concur to the development of the infection.
Collapse
Affiliation(s)
- Thabata L A Duque
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil.,Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Cynthia M Cascabulho
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Gabriel M Oliveira
- Laboratório de Biologia Celular, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
35
|
Hoffman KA, Reynolds C, Bottazzi ME, Hotez P, Jones K. Improved Biomarker and Imaging Analysis for Characterizing Progressive Cardiac Fibrosis in a Mouse Model of Chronic Chagasic Cardiomyopathy. J Am Heart Assoc 2019; 8:e013365. [PMID: 31718442 PMCID: PMC6915297 DOI: 10.1161/jaha.119.013365] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Chronic chagasic cardiomyopathy (CCC), caused by Trypanosoma cruzi infection, is an important public health problem attributable to progressive cardiomyopathy in patients, for which there is no cure. Chronic chagasic cardiomyopathy is characterized by myocarditis and cardiac fibrosis, which leads to life‐threatening arrhythmogenic and circulatory abnormalities. This study aimed to investigate cardiac fibrosis progression in a mouse model of chronic chagasic cardiomyopathy. Methods and Results Cardiac cells infected with T cruzi produced significantly higher concentrations of transforming growth factor‐β (TGF‐β), connective tissue growth factor, endothelin‐1, and platelet‐derived growth factor‐D than noninfected controls. Female Balb/c mice infected with T cruzi were compared with naïve mice. TGF‐β genes and other TGF‐β superfamily genes, as well as connective tissue growth factor, endothelin‐1, and platelet‐derived growth factor, were upregulated in infected mouse hearts. Serum concentrations of TGF‐β, connective tissue growth factor, and platelet‐derived growth factor‐D were higher in infected mice and correlated with cardiac fibrosis. Strain analysis performed on magnetic resonance images at 111 and 140 days postinfection and echocardiography images at 212 days postinfection revealed significantly elevated left ventricular strain and cardiac fibrosis and concomitantly significantly decreased cardiac output in infected mice. Conclusions TGF‐β, connective tissue growth factor and platelet‐derived growth factor‐D are potentially useful biomarkers of cardiac fibrosis, as they correlate with cardiac fibrosis. Strain analysis allows for use of noninvasive methods to measure fibrosis in the chronic stages of chagasic cardiomyopathy in a mouse model. These findings can be applied as noninvasive tools to study the effects of interventions and/or therapeutics on cardiac fibrosis development when using a mouse model of chronic chagasic cardiomyopathy.
Collapse
Affiliation(s)
- Kristyn A Hoffman
- Department of Molecular Virology and Microbiology Baylor College of Medicine Houston TX.,Texas Children's Hospital Center for Vaccine Development Houston TX
| | - Corey Reynolds
- Department of Molecular Physiology Baylor College of Medicine Houston TX
| | - Maria Elena Bottazzi
- Department of Molecular Virology and Microbiology Baylor College of Medicine Houston TX.,Department of Pediatrics Section of Tropical Medicine Baylor College of Medicine Houston TX.,Texas Children's Hospital Center for Vaccine Development Houston TX.,Department of Biology Baylor University Waco TX
| | - Peter Hotez
- Department of Molecular Virology and Microbiology Baylor College of Medicine Houston TX.,Department of Pediatrics Section of Tropical Medicine Baylor College of Medicine Houston TX.,Texas Children's Hospital Center for Vaccine Development Houston TX.,Department of Biology Baylor University Waco TX
| | - Kathryn Jones
- Department of Molecular Virology and Microbiology Baylor College of Medicine Houston TX.,Department of Pediatrics Section of Tropical Medicine Baylor College of Medicine Houston TX.,Texas Children's Hospital Center for Vaccine Development Houston TX
| |
Collapse
|
36
|
Martinez F, Perna E, Perrone SV, Liprandi AS. Chagas Disease and Heart Failure: An Expanding Issue Worldwide. Eur Cardiol 2019; 14:82-88. [PMID: 31360228 PMCID: PMC6659042 DOI: 10.15420/ecr.2018.30.2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/01/2019] [Indexed: 01/28/2023] Open
Abstract
Chagas disease, originally a South American endemic health problem, is expanding worldwide because of people migration. Its main impact is on the cardiovascular system, producing myocardial damage that frequently results in heart failure. Pathogenic pathways are mainly related to inmunoinflamatory reactions in the myocardium and, less frequently, in the gastrointestinal tract. The heart usually shows fibrosis, producing dilatation and damage of the electrogenic cardiac system. These changes result in cardiomyopathy with heart failure and frequent cardiac arrhythmias and heart blocks. Diagnosis of the disease must include a lab test to detect the parasite or its immune reactions and the usual techniques to evaluate cardiac function. Therapeutic management of Chagas heart failure does not differ significantly from the most common treatment for dilated cardiomyopathy, with special focus on arrhythmias and several degrees of heart block. Heart transplantation is reserved for end-stage cases. Major international scientific organisations are delivering recommendations for prevention and early diagnosis. This article provides an analysis of epidemiology, prevention, treatment and the relationship between Chagas disease and heart failure.
Collapse
Affiliation(s)
- Felipe Martinez
- Cordoba National University, Instituto DAMIC Córdoba, Argentina.,Docencia, Asistencia Médica e Investigación Clínica (DAMIC) Medical Institute, Rusculleda Foundation for Research Córdoba Argentina
| | - Eduardo Perna
- Coronary Care Unit and Heart Failure Division, Juana Cabral Cardiovascular Institute Corrientes, Argentina
| | - Sergio V Perrone
- El Cruce Hospital Buenos Aires, Argentina.,Argentine Catholic University Buenos Aires, Argentina
| | - Alvaro Sosa Liprandi
- Cardiovascular Division, Sanatorio Güemes Hospital Buenos Aires, Argentina.,Postgraduate Medical School in Cardiology Universidad de Buenos Aires, Argentina
| |
Collapse
|
37
|
Gupta S, Salgado-Jiménez B, Lokugamage N, Vázquez-Chagoyán JC, Garg NJ. TcG2/TcG4 DNA Vaccine Induces Th1 Immunity Against Acute Trypanosoma cruzi Infection: Adjuvant and Antigenic Effects of Heterologous T. rangeli Booster Immunization. Front Immunol 2019; 10:1456. [PMID: 31293599 PMCID: PMC6606718 DOI: 10.3389/fimmu.2019.01456] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/10/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Chagas cardiomyopathy is caused by Trypanosoma cruzi (Tc). Two antigenic candidates, TcG2 and TcG4, are recognized by antibodies in naturally infected dogs and humans; and these vaccine candidates provided protection from Tc infection in mice and dogs. Trypanosoma rangeli (Tr) is non-pathogenic to mammals and shown to elicit cross-reactive anti-Tc antibodies. In this study, we investigated if fixed Tr (fTr) can further enhance the efficacy of the TcG2/TcG4 DNA vaccine. Methods and Results: C57BL/6 mice were immunized with TcG2/TcG4 DNA vaccine and fTr (delivered as an adjuvant or in prime-boost approach), and challenged with Tc. Serology studies showed that fTr (±quil-A) elicited Tc- and Tr-reactive IgGs that otherwise were not stimulated by TcG2/TcG4 vaccine only, and quil-A had suppressive effects on fTr-induced IgGs. After challenge infection, TcG2/TcG4-vaccinated mice exhibited potent expansion of antigen- and Tc-specific IgGs that were not boosted by fTr±quil-A. Flow cytometry analysis showed that TcG2/TcG4-induced dendritic cells (DC) and macrophages (Mφ) responded to challenge infection by expression of markers of antigen uptake, processing, and presentation, and production of pro-inflammatory cytokines. TcG2/TcG4-induced CD4+T cells acquired Th1 phenotype and expressed markers that orchestrate adaptive immunity. A fraction of vaccine-induced CD4+T cells exhibited iTreg phenotype responsible for aversion of self-injurious immune responses. Further, TcG2/TcG4-vaccinated mice exhibited potent expansion of poly-functional CD8+T cells with TNF-α/IFN-γ production and cytolytic phenotype post-infection. Subsequently, tissue parasites and pathology were hardly detectable in TcG2/TcG4-vaccinated/infected mice. Inclusion of fTr±quil-A had no clear additive effects in improving the Tc-specific adaptive immunity and parasite control than was noted in mice vaccinated with TcG2/TcG4 alone. Non-vaccinated mice lacked sufficient activation of Th1 CD4+/CD8+T cells, and exhibited >10-fold higher levels of tissue parasite burden than was noted in vaccinated/infected mice. Conclusion:TcG2/TcG4 vaccine elicits highly effective immunity, and inclusion of fTr is not required to improve the efficacy of DNA vaccine against acute Tc infection in mice.
Collapse
Affiliation(s)
- Shivali Gupta
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Berenice Salgado-Jiménez
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nandadeva Lokugamage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Juan Carlos Vázquez-Chagoyán
- Facultad de Medicina Veterinaria y Zootecnia, Centro de Investigación y Estudios Avanzados en Salud Animal, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
38
|
Zago MP, Wiktorowicz JE, Spratt H, Koo SJ, Barrientos N, Nuñez Burgos A, Nuñez Burgos J, Iñiguez F, Botelli V, Leon de la Fuente R, Garg NJ. Potential Utility of Protein Targets of Cysteine-S-Nitrosylation in Identifying Clinical Disease Status in Human Chagas Disease. Front Microbiol 2019; 9:3320. [PMID: 30697201 PMCID: PMC6340995 DOI: 10.3389/fmicb.2018.03320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/20/2018] [Indexed: 01/24/2023] Open
Abstract
Trypanosoma cruzi (Tc) infection causes Chagas disease (ChD) presented by dilated cardiomyopathy and heart failure. During infection, oxidative and nitrosative stresses are elicited by the immune cells for control the pathogen; however, excess nitric oxide and superoxide production can result in cysteine S-nitrosylation (SNO) of host proteins that affects cellular homeostasis and may contribute to disease development. To identify the proteins with changes in SNO modification levels as a hallmark of ChD, we obtained peripheral blood mononuclear cells (PBMC) from seronegative, normal healthy (NH, n = 30) subjects, and from seropositive clinically asymptomatic (ChD CA, n = 25) or clinically symptomatic (ChD CS, n = 28) ChD patients. All samples were treated (Asc+) or not-treated (Asc−) with ascorbate (reduces nitrosylated thiols), labeled with the thiol-labeling BODIPY FL-maleimide dye, resolved by two-dimensional electrophoresis (total 166 gels), and the protein spots that yielded significant differences in abundance or SNO level at p-value of ≤ 0.05t−test/Welch/BH were identified by MALDI-TOF/TOF MS or OrbiTrap LC-MS/MS. Targeted analysis of a new cohort of PBMC samples (n = 10–14/group) was conducted to verify the differential abundance/SNO levels of two of the proteins in ChD (vs. NH) subjects. The multivariate adaptive regression splines (MARS) modeling, comparing differences in relative SNO level (Asc−/Asc+ ratio) of the protein spots between any two groups yielded SNO biomarkers that exhibited ≥90% prediction success in classifying ChD CA (582-KRT1 and 884-TPM3) and ChD CS (426-PNP, 582-KRT1, 486-ALB, 662-ACTB) patients from NH controls. Ingenuity Pathway Analysis (IPA) of the SNO proteome dataset normalized to changes in protein abundance suggested the proteins belonging to the signaling networks of cell death and the recruitment and migration of immune cells were most affected in ChD CA and ChD CS (vs. NH) subjects. We propose that SNO modification of the select panel of proteins identified in this study have the potential to identify ChD severity in seropositive individuals exposed to Tc infection.
Collapse
Affiliation(s)
- Maria Paola Zago
- Instituto de Patología Experimental, CONICET-UNSa, Salta, Argentina
| | - John E Wiktorowicz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch (UTMB), Galveston, TX, United States.,Institute for Human Infections and Immunity, University of Texas Medical Branch (UTMB), Galveston, TX, United States
| | - Heidi Spratt
- Department of Preventive Medicine and Community Health, University of Texas Medical Branch (UTMB), Galveston, TX, United States
| | - Sue-Jie Koo
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, United States
| | | | - Aida Nuñez Burgos
- Servicio de Cardiología, Programa de Medicina Interna, Hospital Papa Francisco, Salta, Argentina
| | - Julio Nuñez Burgos
- Servicio de Cardiología, Programa de Medicina Interna, Hospital Papa Francisco, Salta, Argentina
| | - Facundo Iñiguez
- Servicio de Cardiología, Hospital San Bernardo, Salta, Argentina
| | | | | | - Nisha Jain Garg
- Institute for Human Infections and Immunity, University of Texas Medical Branch (UTMB), Galveston, TX, United States.,Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, United States.,Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, TX, United States
| |
Collapse
|
39
|
Bonney KM, Luthringer DJ, Kim SA, Garg NJ, Engman DM. Pathology and Pathogenesis of Chagas Heart Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:421-447. [PMID: 30355152 DOI: 10.1146/annurev-pathol-020117-043711] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chagas heart disease is an inflammatory cardiomyopathy that develops in approximately one-third of people infected with the protozoan parasite Trypanosoma cruzi. One way T. cruzi is transmitted to people is through contact with infected kissing bugs, which are found in much of the Western Hemisphere, including in vast areas of the United States. The epidemiology of T. cruzi and Chagas heart disease and the varied mechanisms leading to myocyte destruction, mononuclear cell infiltration, fibrosis, and edema in the heart have been extensively studied by hundreds of scientists for more than 100 years. Despite this wealth of knowledge, it is still impossible to predict what will happen in an individual infected with T. cruzi because of the tremendous variability in clonal parasite virulence and human susceptibility to infection and the lack of definitive molecular predictors of outcome from either side of the host-parasite equation. Further, while several distinct mechanisms of pathogenesis have been studied in isolation, it is certain that multiple coincident mechanisms combine to determine the ultimate outcome. For these reasons, Chagas disease is best considered a collection of related but distinct illnesses. This review highlights the pathology and pathogenesis of the most common adverse sequela of T. cruzi infection-Chagas heart disease-and concludes with a discussion of key unanswered questions and a view to the future.
Collapse
Affiliation(s)
- Kevin M Bonney
- Liberal Studies, Faculty of Arts and Sciences, New York University, New York, NY 10003, USA;
| | - Daniel J Luthringer
- Department of Pathology and Laboratory Medicine and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA; , ,
| | - Stacey A Kim
- Department of Pathology and Laboratory Medicine and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA; , ,
| | - Nisha J Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555-1070, USA;
| | - David M Engman
- Department of Pathology and Laboratory Medicine and Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA; , ,
| |
Collapse
|
40
|
Wen JJ, Garg NJ. Manganese superoxide dismutase deficiency exacerbates the mitochondrial ROS production and oxidative damage in Chagas disease. PLoS Negl Trop Dis 2018; 12:e0006687. [PMID: 30044789 PMCID: PMC6078326 DOI: 10.1371/journal.pntd.0006687] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 08/06/2018] [Accepted: 07/13/2018] [Indexed: 02/04/2023] Open
Abstract
In this study, we have investigated the effects of manganese superoxide dismutase (SOD2 or MnSOD) deficiency on mitochondrial function and oxidative stress during Chagas disease. For this, C57BL/6 wild type (WT) and MnSOD+/- mice were infected with Trypanosoma cruzi (Tc), and evaluated at 150 days’ post-infection that corresponded to chronic disease phase. Genetic deletion of SOD2 decreased the expression and activity of MnSOD, but it had no effect on the expression of other members of the SOD family. The myocardial expression and activity of MnSOD were significantly decreased in chronically infected WT mice, and it was further worsened in MnSOD+/- mice. Chronic T. cruzi infection led to a decline in mitochondrial complex I and complex II driven, ADP-coupled respiration and ATP synthesis in the myocardium of WT mice. The baseline oxidative phosphorylation (OXPHOS) capacity in MnSOD+/- mice was decreased, and it had an additive effect on mitochondrial dysregulation of ATP synthesis capacity in chagasic myocardium. Further, MnSOD deficiency exacerbated the mitochondrial rate of reactive oxygen species (ROS) production and myocardial oxidative stress (H2O2, protein carbonyls, malondialdehyde, and 4-hydroxynonenal) in Chagas disease. Peripheral and myocardial parasite burden and inflammatory response (myeloperoxidase, IL-6, lactate dehydrogenase, inflammatory infiltrate) were increased in all chagasic WT and MnSOD+/- mice. We conclude that MnSOD deficiency exacerbates the loss in mitochondrial function and OXPHOS capacity and enhances the myocardial oxidative damage in chagasic cardiomyopathy. Mitochondria targeted, small molecule mitigators of MnSOD deficiency will offer potential benefits in averting the mitochondrial dysfunction and chronic oxidative stress in Chagas disease. Infection by Trypanosoma cruzi parasitic protozoan remains endemic in Latin America. After acute parasitemia phase is controlled by host immune system, infected individuals remain clinically silent but manifest a number of micro and macro cardiac injuries for several years. Eventually many of the infected individuals develop chronic cardiomyopathy that leads to heart failure and sudden death. Cardiac muscle cells are rich in mitochondria and manganese superoxide dismutase (MnSOD) is the chief superoxide scavenging enzyme in the mitochondria. In this study, we show that a deficiency of MnSOD exacerbates the T. cruzi induced mitochondrial dysfunction of the electron transport chain and energy production in the heart. Further, MnSOD deficiency resulted in increased mitochondrial release of oxidants and caused excessive oxidative damage in the chagasic heart. Our results suggest that small molecule agonists of MnSOD will have potential utility as adjuvant therapy in preventing the development of chronic Chagas disease in infected individuals.
Collapse
Affiliation(s)
- Jake J. Wen
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas, United States of America
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas, United States of America
- Department of Pathology, UTMB, Galveston, Texas, United States of America
- Institute for Human Infections and Immunity, UTMB, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
41
|
Chagas Cardiomyopathy: Evidence in Medical and Nutritional Management. CURRENT TROPICAL MEDICINE REPORTS 2018. [DOI: 10.1007/s40475-018-0155-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
42
|
González-Alcaide G, Salinas A, Ramos JM. Scientometrics analysis of research activity and collaboration patterns in Chagas cardiomyopathy. PLoS Negl Trop Dis 2018; 12:e0006602. [PMID: 29912873 PMCID: PMC6023249 DOI: 10.1371/journal.pntd.0006602] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/28/2018] [Accepted: 06/10/2018] [Indexed: 12/18/2022] Open
Abstract
Background Chagas cardiomyopathy is a serious and common complication of Chagas disease. Methods Through bibliometric and Social Network Analysis, we examined patterns of research on Chagas cardiomyopathy, identifying the main countries, authors, research clusters, and topics addressed; and measuring the contribution of different countries. Results We found 1932 documents on Chagas cardiomyopathy in the MEDLINE database. The most common document type was ‘journal article’, accounting for 79.6% of the total (n = 1538), followed by ‘review’ (n = 217, 11.2%). The number of published records increased from 156 in 1980–1984 to 311 in 2010–2014. Only 2.5% were clinical trials. Brazil and the USA dominated the research, participating in 53.1% and 25.7%, respectively, of the documents. Other Latin American countries where Chagas is endemic contributed less, with Bolivia, where Chagas disease is most prevalent, producing only 1.8% of the papers. We observed a high rate of domestic collaboration (83.1% of the documents published in 2010–2016) and a lower but significant rate of international collaboration (32.5% in the same time period). Although clinical research dominated overall, the USA, Mexico and several countries in Europe produced a considerable body of basic research on animal models. We identified four main research clusters, focused on heart failure and dysfunction (physical symptoms, imaging techniques, treatment), and on myocarditis and parasitemia in animal models. Conclusions Research on Chagas cardiomyopathy increased over the study period. There were more clinical than basic studies, though very few of the documents were clinical trials. Brazil and the USA are currently leading the research on this subject, while some highly endemic countries, such as Bolivia, have contributed very little. Different approaches could help to redress this imbalance: encouraging researchers to conduct more clinical trials, launching international collaborations to help endemic countries contribute more, and strengthening links between basic and clinical research. Scientific production on Chagas cardiomyopathy has grown considerably since the turn of the 21st century, probably reflecting the increased incidence of Chagas disease in non-endemic areas like the USA and Europe. Brazil and the USA dominate the research, but we found a very small proportion of clinical trials on Chagas cardiomyopathy and a low scientific production in several endemic countries with a high prevalence of the disease such as Colombia, Chile, Mexico and Bolivia. We observed a polarity between endemic and non-endemic countries where clinical research and basic research predominate, respectively. Different approaches could help to redress the observed imbalance of research on Chagas cardiomyopathy: encouraging researchers to conduct more clinical trials, launching international collaborations to help endemic countries contribute more, and strengthening links between basic and clinical research. It is crucial to foster translational research in order to link basic knowledge on the physiology of the disease with clinical applications in diagnosis and treatment.
Collapse
Affiliation(s)
- Gregorio González-Alcaide
- Department of History of Science and Documentation, University of Valencia, Valencia, Spain
- * E-mail:
| | | | - José M. Ramos
- Department of Clinical Medicine, Miguel Hernández University of Elche, Alicante, Spain
- Department of Internal Medicine, Hospital General Universitario de Alicante, Alicante, Spain
| |
Collapse
|
43
|
Roscoe A, Tomey MI, Torregrossa G, Galhardo C, Parhar K, Zochios V. Chagas Cardiomyopathy: A Comprehensive Perioperative Review. J Cardiothorac Vasc Anesth 2018; 32:2780-2788. [PMID: 29803311 DOI: 10.1053/j.jvca.2018.04.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Andrew Roscoe
- Department of Cardiothoracic Anesthesia and Critical Care Medicine, Papworth Hospital, Cambridge, United Kingdom
| | - Matthew I Tomey
- Cardiovascular Institute and Institute for Critical Care Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gianluca Torregrossa
- Department of Cardiothoracic Surgery, Mount Sinai Saint Luke, Mount Sinai Health System, New York, NY
| | - Carlos Galhardo
- Department of Anesthesia, National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Ken Parhar
- Department of Critical Care Medicine, University of Calgary, Calgary, Canada
| | - Vasileios Zochios
- University Hospitals Birmingham NHS Foundation Trust, Department of Critical Care Medicine, Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham, United Kingdom; Perioperative Critical Care and Trauma Trials Group, Institute of Inflammation and Ageing, Centre of Translational Inflammation Research, University of Birmingham, Birmingham, United Kingdom.
| |
Collapse
|
44
|
Trypanosoma cruzi Produces the Specialized Proresolving Mediators Resolvin D1, Resolvin D5, and Resolvin E2. Infect Immun 2018; 86:IAI.00688-17. [PMID: 29358332 DOI: 10.1128/iai.00688-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/14/2018] [Indexed: 12/31/2022] Open
Abstract
Trypanosoma cruzi is a protozoan parasite that causes Chagas disease (CD). CD is a persistent, lifelong infection affecting many organs, most notably the heart, where it may result in acute myocarditis and chronic cardiomyopathy. The pathological features include myocardial inflammation and fibrosis. In the Brazil strain-infected CD-1 mouse, which recapitulates many of the features of human infection, we found increased plasma levels of resolvin D1 (RvD1), a specialized proresolving mediator of inflammation, during both the acute and chronic phases of infection (>100 days postinfection) as determined by enzyme-linked immunosorbent assay (ELISA). Additionally, ELISA on lysates of trypomastigotes of both strains Tulahuen and Brazil revealed elevated levels of RvD1 compared with lysates of cultured epimastigotes of T. cruzi, tachyzoites of Toxoplasma gondii, trypomastigotes of Trypanosoma brucei, cultured L6E9 myoblasts, and culture medium containing no cells. Lysates of T. cruzi-infected myoblasts also displayed increased levels of RvD1. Lipid mediator metabolomics confirmed that the trypomastigotes of T. cruzi produced RvD1, RvD5, and RvE2, which have been demonstrated to modulate the host response to bacterial infections. Plasma RvD1 levels may be both host and parasite derived. Since T. cruzi synthesizes specialized proresolving mediators of inflammation, as well as proinflammatory eicosanoids, such as thromboxane A2, one may speculate that by using these lipid mediators to modulate its microenvironment, the parasite is able to survive.
Collapse
|
45
|
Lopez M, Tanowitz HB, Garg NJ. Pathogenesis of Chronic Chagas Disease: Macrophages, Mitochondria, and Oxidative Stress. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018; 5:45-54. [PMID: 29868332 PMCID: PMC5983038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
PURPOSE OF REVIEW Trypanosoma cruzi is the causative agent of Chagas disease. Decades after initial infection, ~30% of individuals can develop chronic chagasic cardiomyopathy. There are several proposed mechanisms for pathogenesis of Chagas disease, including parasite persistence, immune responses against parasite or self that continue in the heart, vascular compromise, and involvement of autonomous and central nervous system. Herein, we will focus on the significance of macrophages, mitochondrial dysfunction, and oxidative stress in progression of chagasic cardiomyopathy. RECENT FINDINGS The current literature suggests that T. cruzi prevents cytotoxic activities of the innate immune cells and persists in the host, contributing to mitochondrial oxidative stress. We discuss how the neoantigens generated due to cellular oxidative damage contribute to chronic inflammatory stress in chagasic disease. SUMMARY We propose that metabolic regulators, PARP-1/SIRT1, determine the disease outcome by modulating the mitochondrial and macrophage stress and antioxidant/oxidant imbalance, and offer a potential new therapy against chronic Chagas disease.
Collapse
Affiliation(s)
- Marcos Lopez
- Translational Biomedical Research Group, Fundación Cardiovascular de Colombia, Floridablanca, Colombia and Graduate Program in Biomedical Sciencies, Faculty of Health, Universidad del Valle, Cali, Colombia
| | - Herbert B Tanowitz
- Departments of Pathology and Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Nisha J Garg
- Departments of Microbiology and Immunology and Pathology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas 77555-1070
| |
Collapse
|
46
|
Lopez M, Tanowitz HB, Garg NJ. Pathogenesis of Chronic Chagas Disease: Macrophages, Mitochondria, and Oxidative Stress. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018. [DOI: 10.1007/s40588-018-0081-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
47
|
Mascareno E, Gupta R, Martello LA, Dhar-Mascareno M, Salciccioli L, Beckles D, Walsh MG, Machado FS, Tanowitz HB, Haseeb M. Rapidly progressive course of Trypanosoma cruzi infection in mice heterozygous for hexamethylene bis-acetamide inducible 1 (Hexim1) gene. Microbes Infect 2018; 20:25-36. [DOI: 10.1016/j.micinf.2017.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/30/2017] [Accepted: 09/04/2017] [Indexed: 01/02/2023]
|
48
|
Chronic Chagas cardiomyopathy: a therapeutic challenge and future strategies. Emerg Top Life Sci 2017; 1:579-584. [PMID: 33525838 DOI: 10.1042/etls20170109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/19/2017] [Accepted: 10/24/2017] [Indexed: 11/17/2022]
Abstract
Infectious diseases are the main cause of acquired dilated cardiomyopathy. This group of disorders shares in common inflammatory cell infiltrate and myocardial remodeling. As part of its pathophysiology, there is coronary microvascular dysfunction, distinct from that observed in coronary artery disease. Chagas cardiomyopathy presents several vascular characteristics that are similar to those presented in other acquired cardiomyopathies. There is convincing evidence of the microvascular involvement and the inflammatory processes that lead to endothelial activation and ischemic damage. Current therapy for the Chagas disease is limited, and it is proposed to combine it with other pharmacological strategies that modify critical physiopathological aspects beneficial for the clinical course of the Chagas cardiomyopathy.
Collapse
|
49
|
Integration of miRNA and gene expression profiles suggest a role for miRNAs in the pathobiological processes of acute Trypanosoma cruzi infection. Sci Rep 2017; 7:17990. [PMID: 29269773 PMCID: PMC5740174 DOI: 10.1038/s41598-017-18080-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/01/2017] [Indexed: 12/20/2022] Open
Abstract
Chagas disease, caused by the parasite Trypanosoma cruzi, is endemic in Latin America. Its acute phase is associated with high parasitism, myocarditis and profound myocardial gene expression changes. A chronic phase ensues where 30% develop severe heart lesions. Mouse models of T. cruzi infection have been used to study heart damage in Chagas disease. The aim of this study was to provide an interactome between miRNAs and their targetome in Chagas heart disease by integrating gene and microRNA expression profiling data from hearts of T. cruzi infected mice. Gene expression profiling revealed enrichment in biological processes and pathways associated with immune response and metabolism. Pathways, functional and upstream regulator analysis of the intersections between predicted targets of differentially expressed microRNAs and differentially expressed mRNAs revealed enrichment in biological processes and pathways such as IFNγ, TNFα, NF-kB signaling signatures, CTL-mediated apoptosis, mitochondrial dysfunction, and Nrf2-modulated antioxidative responses. We also observed enrichment in other key heart disease-related processes like myocarditis, fibrosis, hypertrophy and arrhythmia. Our correlation study suggests that miRNAs may be implicated in the pathophysiological processes taking place the hearts of acutely T. cruzi-infected mice.
Collapse
|
50
|
Elucidating the impact of low doses of nano-formulated benznidazole in acute experimental Chagas disease. PLoS Negl Trop Dis 2017; 11:e0006119. [PMID: 29267280 PMCID: PMC5755931 DOI: 10.1371/journal.pntd.0006119] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 01/05/2018] [Accepted: 11/17/2017] [Indexed: 01/05/2023] Open
Abstract
Background Chagas disease is a neglected parasitic infection caused by the protozoan Trypanosoma cruzi (T. cruzi) that affects more than 6 million people, mainly in Latin America. Benznidazole is still the drug of choice in many countries to treat it in spite of its dosage regimen and adverse side effects such as such as allergic dermatitis, peripheral neuropathy and anorexia. Thus, novel, safer, and more efficacious treatments for such neglected infection are urgently required. Methodology In this study, the efficacy of orally administered low doses of benznidazole (BNZ) nanoparticles was evaluated during the acute phase in mice infected with T. cruzi Nicaragua (TcN) that were immunosuppressed during the chronic stage of the disease. Moreover, the production of T. cruzi-specific antibodies, cardiac tissue inflammation and reactive oxygen species generation by Vero cells treated with both BNZ nanoparticles (BNZ-nps) and raw BNZ (R-BNZ) were also evaluated. Principal findings T. cruzi infected mice treated with 10, 25 or 50 mg/kg/day of BNZ-nps survived until euthanasia (92 days post infection (dpi)), while only 15% of infected untreated mice survived until the end of the experiment. PCR analysis of blood samples taken after induction of immunosuppression showed that a dosage of 25 mg/kg/day rendered 40% of the mice PCR-negative. The histological analysis of heart tissue showed a significant decrease in inflammation after treatments with 25 and 50 mg/kg/day, while a similar inflammatory damage was observed in both infected mice treated with R-BNZ (50 mg/kg/day) and untreated mice. In addition, only BNZ-nps treated mice led to lower levels of T. cruzi-specific antibodies to 50–100%. Finally, mammalian Vero cells treated with BNZ-nps or R-BNZ lead to a significant increase in ROS production. Conclusions Based on these findings, this research highlights the in-vitro/in-vivo efficacy of nanoformulated BNZ against T. cruzi acute infections in immunosuppressed and non-immunosuppressed mice and provides further evidence for the optimization of dosage regimens to treat Chagas disease. Chagas disease is a neglected parasitic infection caused by the protozoan Trypanosoma cruzi (T. cruzi) that affects more than 6 million people, mainly in Latin America. Benznidazole is still the drug of choice in many countries to treat it in spite of its dosage regimen and adverse side effects such as such as allergic dermatitis, peripheral neuropathy and anorexia. In this study, the efficacy of low doses of benznidazole, formulated as nanoparticles, against T. cruzi acute infections in immunosuppressed and non-immunosuppressed mice was investigated in order to establish future treatment strategies. In-vivo experiments showed that all infected mice treated with low doses of nanoformulated benznidazole survived until the end of the assay (92 dpi), while only 15% of infected untreated mice survived to the end of the same period of time. Moreover, such novel formulation was able to decrease the parasite burden and, consequently, heart inflammation and lesions were significantly reduced. Clearly, low doses of benznidazole exhibited, at least, the same efficacy in infected mice as the usual dose, confirming the usefulness of nanoformulated benznidazole for an improved treatment of Chagas disease.
Collapse
|