1
|
Ricci MF, Lourenço EMG, Pereira RDD, Araújo RRS, Oliveira FBR, Barbosa da Silva E, de Oliveira GS, Teixeira MM, Rocha NDN, Chambergo FS, Roman-Campos D, Cruz JS, Ferreira RS, Machado FS. Zileuton, a 5-Lypoxigenase Inhibitor, is Antiparasitic and Prevents Inflammation in the Chronic Stage of Heart Chagas Disease. ACS Infect Dis 2024; 10:4258-4270. [PMID: 39609255 DOI: 10.1021/acsinfecdis.4c00623] [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: 11/30/2024]
Abstract
Chronic Chagas cardiomyopathy is associated with an unbalanced immune response and impaired heart function, and available drugs do not prevent its development. Zileuton (Zi), a 5-lypoxigenase inhibitor, affects inflammatory/pro-resolution mediators. Herein, Zi treatment in the early phase of infection reduced parasitemia associated mainly with the direct effect of Zi on the parasite, and the enzyme epoxide hydrolase was the potential molecular target behind the trypanocidal effect. In the intermediate acute phase of infection, Zi reduced the number of innate and adaptive inflammatory cells, increased the level of SOCS2 expression in the heart associated with lower inflammation, and improved cardiac function. Zi treatment initiated in the chronic stage increased the level of SOCS2 expression in the heart, reduced inflammation, and improved cardiac function. Our data suggest that Zi protects against Trypanosoma cruzi infection by acting directly on the parasite and reducing heart damage and is a promising option for the treatment of Chagas disease.
Collapse
Affiliation(s)
- Mayra Fernanda Ricci
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Estela Mariana Guimarães Lourenço
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Rafaela das Dores Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Ronan Ricardo Sabino Araújo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Fernando Bento Rodrigues Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Elany Barbosa da Silva
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Gabriel Stephani de Oliveira
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, 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 30130-100, Minas Gerais, Brazil
| | - Nazareth de Novaes Rocha
- Department of Physiology and Pharmacology, Biomedical Institute, Universidade Federal Fluminense, Niterói 24020-141, Rio de Janeiro, Brazil
| | | | - Danilo Roman-Campos
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Jader Santos Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Rafaela Salgado Ferreira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, 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 30130-100, Minas Gerais, Brazil
| |
Collapse
|
2
|
Nicolau ST, Tres DP, Ayala TS, Menolli RA. Nonsteroidal Anti-Inflammatory Drugs and Experimental Chagas Disease: An Unsolved Question. Parasite Immunol 2024; 46:e13057. [PMID: 39008292 DOI: 10.1111/pim.13057] [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] [Received: 12/08/2023] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
Chagas disease is a parasitic disease caused by the protozoan Trypanosoma cruzi with an acute, detectable blood parasites phase and a chronic phase, in which the parasitemia is not observable, but cardiac and gastrointestinal consequences are possible. Mice are the principal host used in experimental Chagas disease but reproduce the human infection depending on the animal and parasite strain, besides dose and route of administration. Lipidic mediators are tremendously involved in the pathogenesis of T. cruzi infection, meaning the prostaglandins and thromboxane, which participate in the immunosuppression characteristic of the acute phase. Thus, the eicosanoids inhibition caused by the nonsteroidal anti-inflammatory drugs (NSAIDs) alters the dynamic of the disease in the experimental models, both in vitro and in vivo, which can explain the participation of the different mediators in infection. However, marked differences are founded in the various NSAIDs existing because of the varied routes blocked by the drugs. So, knowing the results in the experimental models of Chagas disease with or without the NSAIDs helps comprehend the pathogenesis of this infection, which still needs a better understanding.
Collapse
Affiliation(s)
- Scheila Thaís Nicolau
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| | - Daniela Patrícia Tres
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| | - Thaís Soprani Ayala
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| | - Rafael Andrade Menolli
- Laboratory of Applied Immunology, Center of Medical and Pharmaceutical Sciences, Western Parana State University, Cascavel, Brazil
| |
Collapse
|
3
|
Nisimura LM, Ferreira RR, Coelho LL, de Oliveira GM, Gonzaga BM, Meuser-Batista M, Lannes-Vieira J, Araujo-Jorge T, Garzoni LR. Vascular Growth Factor Inhibition with Bevacizumab Improves Cardiac Electrical Alterations and Fibrosis in Experimental Acute Chagas Disease. BIOLOGY 2023; 12:1414. [PMID: 37998013 PMCID: PMC10669550 DOI: 10.3390/biology12111414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 11/25/2023]
Abstract
Chagas disease (CD) caused by Trypanosoma cruzi is a neglected illness and a major reason for cardiomyopathy in endemic areas. The existing therapy generally involves trypanocidal agents and therapies that control cardiac alterations. However, there is no treatment for the progressive cardiac remodeling that is characterized by inflammation, microvasculopathy and extensive fibrosis. Thus, the search for new therapeutic strategies aiming to inhibit the progression of cardiac injury and failure is necessary. Vascular Endothelial Growth Factor A (VEGF-A) is the most potent regulator of vasculogenesis and angiogenesis and has been implicated in inducing exacerbated angiogenesis and fibrosis in chronic inflammatory diseases. Since cardiac microvasculopathy in CD is also characterized by exacerbated angiogenesis, we investigated the effect of inhibition of the VEGF signaling pathway using a monoclonal antibody (bevacizumab) on cardiac remodeling and function. Swiss Webster mice were infected with Y strain, and cardiac morphological and molecular analyses were performed. We found that bevacizumab significantly increased survival, reduced inflammation, improved cardiac electrical function, diminished angiogenesis, decreased myofibroblasts in cardiac tissue and restored collagen levels. This work shows that VEGF is involved in cardiac microvasculopathy and fibrosis in CD and the inhibition of this factor could be a potential therapeutic strategy for CD.
Collapse
Affiliation(s)
- Lindice Mitie Nisimura
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.M.N.); (R.R.F.); (L.L.C.); (B.M.G.); (M.M.-B.); (T.A.-J.)
| | - Roberto Rodrigues Ferreira
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.M.N.); (R.R.F.); (L.L.C.); (B.M.G.); (M.M.-B.); (T.A.-J.)
- Laboratory of Applied Genomics and Bioinnovations, Oswaldo Cruz Institute (LAGABI-IOC/Fiocruz), Rio de Janeiro 21040-900, Brazil
| | - Laura Lacerda Coelho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.M.N.); (R.R.F.); (L.L.C.); (B.M.G.); (M.M.-B.); (T.A.-J.)
| | - Gabriel Melo de Oliveira
- Laboratory of Cell Biology, Oswaldo Cruz Institute (LBC-IOC/Fiocruz), Rio de Janeiro 21040-900, Brazil;
| | - Beatriz Matheus Gonzaga
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.M.N.); (R.R.F.); (L.L.C.); (B.M.G.); (M.M.-B.); (T.A.-J.)
| | - Marcelo Meuser-Batista
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.M.N.); (R.R.F.); (L.L.C.); (B.M.G.); (M.M.-B.); (T.A.-J.)
| | - Joseli Lannes-Vieira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute (LBI-IOC/Fiocruz), Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil;
| | - Tania Araujo-Jorge
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.M.N.); (R.R.F.); (L.L.C.); (B.M.G.); (M.M.-B.); (T.A.-J.)
| | - Luciana Ribeiro Garzoni
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Av. Brasil, 4365, Manguinhos, Rio de Janeiro 21040-900, Brazil; (L.M.N.); (R.R.F.); (L.L.C.); (B.M.G.); (M.M.-B.); (T.A.-J.)
| |
Collapse
|
4
|
dos Santos LF, Rodrigues GF, Malvezi AD, de Souza M, Nakama RP, Lovo-Martins MI, Pinge-Filho P. Beneficial effects of acetylsalicylic acid (aspirin) on the actions of extracellular vesicles shed by Trypanosoma cruzi in macrophages. Parasitol Int 2023; 92:102697. [DOI: 10.1016/j.parint.2022.102697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
|
5
|
Díaz-Viraqué F, Chiribao ML, Paes-Vieira L, Machado MR, Faral-Tello P, Tomasina R, Trochine A, Robello C. New Insights into the Role of the Trypanosoma cruzi Aldo-Keto Reductase TcAKR. Pathogens 2023; 12:pathogens12010085. [PMID: 36678433 PMCID: PMC9860839 DOI: 10.3390/pathogens12010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Chagas disease is a zoonotic infectious disease caused by the protozoan parasite Trypanosoma cruzi. It is distributed worldwide, affecting around 7 million people; there is no effective treatment, and it constitutes a leading cause of disability and premature death in the Americas. Only two drugs are currently approved for the treatment, Benznidazole and Nifurtimox, and both have to be activated by reducing the nitro-group. The T. cruzi aldo-keto reductase (TcAKR) has been related to the metabolism of benznidazole. TcAKR has been extensively studied, being most efforts focused on characterizing its implication in trypanocidal drug metabolism; however, little is known regarding its biological role. Here, we found that TcAKR is confined, throughout the entire life cycle, into the parasite mitochondria providing new insights into its biological function. In particular, in epimastigotes, TcAKR is associated with the kinetoplast, which suggests additional roles of the protein. The upregulation of TcAKR, which does not affect TcOYE expression, was correlated with an increase in PGF2α, suggesting that this enzyme is related to PGF2α synthesis in T. cruzi. Structural analysis showed that TcAKR contains a catalytic tetrad conserved in the AKR superfamily. Finally, we found that TcAKR is also involved in Nfx metabolization.
Collapse
Affiliation(s)
- Florencia Díaz-Viraqué
- Laboratorio de Interacciones Hospedero Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - María Laura Chiribao
- Laboratorio de Interacciones Hospedero Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Departamento de Bioquímica, Facultad de Medicina Universidad de la República, Montevideo 11400, Uruguay
| | - Lisvane Paes-Vieira
- Laboratorio de Interacciones Hospedero Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Matias R. Machado
- Unidad de Proteínas Recombinantes, Institut Pasteur de Montevideo, Montevideo 11300, Uruguay
| | - Paula Faral-Tello
- Laboratorio de Interacciones Hospedero Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
| | - Ramiro Tomasina
- Laboratory of Apicomplexan Biology, Institut Pasteur de Montevideo and Departamento de Parasitología, Facultad de Medicina Universidad de la República, Montevideo 11300, Uruguay
| | - Andrea Trochine
- Centro de Referencia en Levaduras y Tecnología Cervecera (CRELTEC), Instituto Andino Patagónico de Tecnologías Biológicas y Geoambientales (IPATEC), CONICET-Universidad Nacional del Comahue, Quintral 1250, San Carlos de Bariloche 8400, Argentina
| | - Carlos Robello
- Laboratorio de Interacciones Hospedero Patógeno, Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay
- Departamento de Bioquímica, Facultad de Medicina Universidad de la República, Montevideo 11400, Uruguay
- Correspondence:
| |
Collapse
|
6
|
Montalvo-Ocotoxtle IG, Rojas-Velasco G, Rodríguez-Morales O, Arce-Fonseca M, Baeza-Herrera LA, Arzate-Ramírez A, Meléndez-Ramírez G, Manzur-Sandoval D, Lara-Romero ML, Reyes-Ortega A, Espinosa-González P, Palacios-Rosas E. Chagas Heart Disease: Beyond a Single Complication, from Asymptomatic Disease to Heart Failure. J Clin Med 2022; 11:7262. [PMID: 36555880 PMCID: PMC9784121 DOI: 10.3390/jcm11247262] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Chagas cardiomyopathy (CC), caused by the protozoan Trypanosoma cruzi, is an important cause of cardiovascular morbidity and mortality in developing countries. It is estimated that 6 to 7 million people worldwide are infected, and it is predicted that it will be responsible for 200,000 deaths by 2025. The World Health Organization (WHO) considers Chagas disease (CD) as a Neglected Tropical Disease (NTD), which must be acknowledged and detected in time, as it remains a clinical and diagnostic challenge in both endemic and non-endemic regions and at different levels of care. The literature on CC was analyzed by searching different databases (Medline, Cochrane Central, EMBASE, PubMed, Google Scholar, EBSCO) from 1968 until October 2022. Multicenter and bioinformatics trials, systematic and bibliographic reviews, international guidelines, and clinical cases were included. The reference lists of the included papers were checked. No linguistic restrictions or study designs were applied. This review is intended to address the current incidence and prevalence of CD and to identify the main pathogenic mechanisms, clinical presentation, and diagnosis of CC.
Collapse
Affiliation(s)
- Isis G. Montalvo-Ocotoxtle
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Gustavo Rojas-Velasco
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Olivia Rodríguez-Morales
- Department of Molecular Biology, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Minerva Arce-Fonseca
- Department of Molecular Biology, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Luis A. Baeza-Herrera
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Arturo Arzate-Ramírez
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Gabriela Meléndez-Ramírez
- Magnetic Resonance Imaging Department, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Daniel Manzur-Sandoval
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Mayra L. Lara-Romero
- Academic Department of Health Sciences, School of Sciences, Universidad de las Américas Puebla, Ex Hacienda Sta. Catarina Mártir S/N. San Andrés Cholula, Puebla 72810, Mexico
| | - Antonio Reyes-Ortega
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Patricia Espinosa-González
- Cardiovascular Critical Care Unit, National Institute of Cardiology “Ignacio Chávez”, Juan Badiano No. 1, Col. Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Erika Palacios-Rosas
- Academic Department of Health Sciences, School of Sciences, Universidad de las Américas Puebla, Ex Hacienda Sta. Catarina Mártir S/N. San Andrés Cholula, Puebla 72810, Mexico
| |
Collapse
|
7
|
Nisimura LM, Ferreira RR, Coelho LL, de Souza EM, Gonzaga BM, Ferrão PM, Waghabi MC, de Mesquita LB, Pereira MCDS, Moreira ODC, Lannes-Vieira J, Garzoni LR. Increased angiogenesis parallels cardiac tissue remodelling in experimental acute Trypanosoma cruzi infection. Mem Inst Oswaldo Cruz 2022; 117:e220005. [PMID: 36417626 PMCID: PMC9677593 DOI: 10.1590/0074-02760220005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Angiogenesis has been implicated in tissue injury in several noninfectious diseases, but its role in Chagas disease (CD) physiopathology is unclear. OBJECTIVES The present study aimed to investigate the effect of Trypanosoma cruzi infection on cardiac angiogenesis during the acute phase of experimental CD. METHODS The signalling pathway involved in blood vessel formation and cardiac remodelling was evaluated in Swiss Webster mice infected with the Y strain of T. cruzi. The levels of molecules involved in the regulation of angiogenesis, such as vascular endothelial growth factor-A (VEGF-A), Flk-1, phosphorylated extracellular-signal-regulated protein kinase (pERK), hypoxia-inducible factor-1α (HIF-1α), CD31, α-smooth muscle actin (α-SMA) and also the blood vessel growth were analysed during T. cruzi infection. Hearts were analysed using conventional histopathology, immunohistochemistry and western blotting. FINDINGS In this study, our data demonstrate that T. cruzi acute infection in mice induces exacerbated angiogenesis in the heart and parallels cardiac remodelling. In comparison with noninfected controls, the cardiac tissue of T. cruzi-infected mice presented higher levels of (i) HIF-1α, VEGF-A, Flk-1 and pERK; (ii) angiogenesis; (iii) α-SMA+ cells in the tissue; and (iv) collagen -1 deposition around blood vessels and infiltrating throughout the myocardium. MAIN CONCLUSIONS We observed cardiac angiogenesis during acute experimental T. cruzi infection parallels cardiac inflammation and remodelling.
Collapse
Affiliation(s)
- Lindice Mitie Nisimura
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil
| | - Roberto Rodrigues Ferreira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil,Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Rio de Janeiro, RJ, Brasil
| | - Laura Lacerda Coelho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil
| | - Elen Mello de Souza
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brasil
| | - Beatriz Matheus Gonzaga
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil
| | - Patrícia Mello Ferrão
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil,Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Genômica Funcional e Bioinformática, Rio de Janeiro, RJ, Brasil
| | - Mariana Caldas Waghabi
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brasil
| | - Liliane Batista de Mesquita
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Ultraestrutura Celular, Rio de Janeiro, RJ, Brasil
| | | | - Otacilio da Cruz Moreira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Biologia Molecular e Doenças Endêmicas, Rio de Janeiro, RJ, Brasil
| | - Joseli Lannes-Vieira
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Biologia das Interações, Rio de Janeiro, RJ, Brasil
| | - Luciana Ribeiro Garzoni
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Rio de Janeiro, RJ, Brasil,+ Corresponding author:
| |
Collapse
|
8
|
Volpini X, Natali L, Brugo MB, de la Cruz-Thea B, Baigorri RE, Cerbán FM, Fozzatti L, Motran CC, Musri MM. Trypanosoma cruzi Infection Promotes Vascular Remodeling and Coexpression of α-Smooth Muscle Actin and Macrophage Markers in Cells of the Aorta. ACS Infect Dis 2022; 8:2271-2290. [PMID: 36083791 DOI: 10.1021/acsinfecdis.2c00318] [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: 01/29/2023]
Abstract
Chagas disease is an emerging global health problem; however, it remains neglected. Increased aortic stiffness (IAS), a predictor of cardiovascular events, has recently been reported in asymptomatic chronic Chagas patients. After vascular injury, smooth muscle cells (SMCs) can undergo alterations associated with phenotypic switch and transdifferentiation, promoting vascular remodeling and IAS. By studying different mouse aortic segments, we tested the hypothesis that Trypanosoma cruzi infection promotes vascular remodeling. Interestingly, the thoracic aorta was the most affected by the infection. Decreased expression of SMC markers and increased expression of proliferative markers were observed in the arteries of acutely infected mice. In acutely and chronically infected mice, we observed cells coexpressing SMC and macrophage (Mo) markers in the media and adventitia layers of the aorta, indicating that T. cruzi might induce cellular processes associated with SMC transdifferentiation into Mo-like cells or vice versa. In the adventitia, the Mo cell functional polarization was associated with an M2-like CD206+arginase-1+ phenotype despite the T. cruzi presence in the tissue. Only Mo-like cells in inflammatory foci were CD206+iNOS+. In addition to the disorganization of elastic fibers, we found thickening of the aortic layers during the acute and chronic phases of the disease. Our findings indicate that T. cruzi infection induces a vascular remodeling with SMC dedifferentiation and increased cell populations coexpressing α-SMA and Mo markers that could be associated with IAS promotion. These data highlight the importance of studying large vessel homeostasis in Chagas disease.
Collapse
Affiliation(s)
- Ximena Volpini
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Lautaro Natali
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Maria Belén Brugo
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Benjamin de la Cruz-Thea
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina
| | - Ruth Eliana Baigorri
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Fabio Marcelo Cerbán
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Laura Fozzatti
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Claudia Cristina Motran
- Centro de Investigaciones en Bioquímica Clínica e Inmunología. Consejo Nacional de Investigaciones Científicas y Técnicas (CIBICI-CONICET), Haya de la Torre y Medina Allende. Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina.,Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba (FCQ-UNC). Ciudad Universitaria, Córdoba, PC X5000HUA, Argentina
| | - Melina Mara Musri
- Instituto de Investigaciones Médicas Mercedes y Martín Ferreyra. Consejo Nacional de Investigaciones Científicas y Tecnicas. Universidad Nacional de Córdoba (INIMEC-CONICET-UNC), Friuli 2434. Colinas de Velez Sarfield, Córdoba, PC X5016NST, Argentina.,Departamento de Fisiología, Facultad de Ciencias Exactas Físicas y Naturales. Universidad Nacional de Córdoba (FCEFyN-UNC). Av. Velez Sarfield 299, Centro, Córdoba, PC X5000JJC, Argentina
| |
Collapse
|
9
|
Rigazio CS, Mariz-Ponte N, Caballero EP, Penas FN, Goren NB, Santamaría MH, Corral RS. Involvement of glycoinositolphospholipid from Trypanosoma cruzi and macrophage migration inhibitory factor in proinflammatory mechanisms promoting cardiovascular injury mechanisms promoting cardiovascular inflammation tThe combined action of glycoinositolphospholipid from Trypanosoma cruzi and macrophage migration inhibitory factor increases proinflammatory mediator production by cardiomyocytes and vascular endothelial cells. Microb Pathog 2022; 173:105881. [DOI: 10.1016/j.micpath.2022.105881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022]
|
10
|
Santi AMM, Ribeiro JM, Reis-Cunha JL, Burle-Caldas GDA, Santos IFM, Silva PA, Resende DDM, Bartholomeu DC, Teixeira SMR, Murta SMF. Disruption of multiple copies of the Prostaglandin F2alpha synthase gene affects oxidative stress response and infectivity in Trypanosoma cruzi. PLoS Negl Trop Dis 2022; 16:e0010845. [PMID: 36260546 PMCID: PMC9581433 DOI: 10.1371/journal.pntd.0010845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/26/2022] [Indexed: 11/08/2022] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, is a serious chronic parasitic disease, currently treated with Nifurtimox (NFX) and Benznidazole (BZ). In addition to high toxicity, these drugs have low healing efficacy, especially in the chronic phase of the disease. The existence of drug-resistant T. cruzi strains and the occurrence of cross-resistance between BZ and NFX have also been described. In this context, it is urgent to study the metabolism of these drugs in T. cruzi, to better understand the mechanisms of resistance. Prostaglandin F2α synthase (PGFS) is an enzyme that has been correlated with parasite resistance to BZ, but the mechanism by which resistance occurs is still unclear. Our results show that the genome of the CL Brener clone of T. cruzi, contains five PGFS sequences and three potential pseudogenes. Using CRISPR/Cas9 we generated knockout cell lines in which all PGFS sequences were disrupted, as shown by PCR and western blotting analyses. The PGFS deletion did not alter the growth of the parasites or their susceptibility to BZ and NFX when compared to wild-type (WT) parasites. Interestingly, NTR-1 transcripts were shown to be upregulated in ΔPGFS mutants. Furthermore, the ΔPGFS parasites were 1.6 to 1.7-fold less tolerant to oxidative stress generated by menadione, presented lower levels of lipid bodies than the control parasites during the stationary phase, and were less infective than control parasites.
Collapse
Affiliation(s)
- Ana Maria Murta Santi
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Juliana Martins Ribeiro
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - João Luís Reis-Cunha
- Departamento de Parasitologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Medicina Veterinária Preventiva, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Paula Alves Silva
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela de Melo Resende
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Silvane Maria Fonseca Murta
- Grupo Genômica Funcional de Parasitos, Instituto René Rachou, Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
11
|
Choudhuri S, Garg NJ. Platelets, Macrophages, and Thromboinflammation in Chagas Disease. J Inflamm Res 2022; 15:5689-5706. [PMID: 36217453 PMCID: PMC9547606 DOI: 10.2147/jir.s380896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
Chagas disease (CD) is a major health problem in the Americas and an emerging health problem in Europe and other nonendemic countries. Several studies have documented persistence of the protozoan parasite Trypanosoma cruzi, and oxidative and inflammatory stress are major pathogenic factor. Mural and cardiac thrombi, cardiac arrhythmias, and cardiomyopathy are major clinical features of CD. During T. cruzi infection, parasite-released factors induce endothelial dysfunction along with platelet (PLT) and immune-cell activation. PLTs have a fundamental role in maintaining hemostasis and preventing bleeding after vascular injury. Excessive activation of PLTs and coagulation cascade can result in thrombosis and thromboembolic events, which are recognized to occur in seropositive individuals in early stages of CD when clinically symptomatic heart disease is not apparent. Several host and parasite factors have been identified to signal hypercoagulability and increase the risk of ischemic stroke in early phases of CD. Further, PLT interaction with immune cells and their role in host defense against pathogens and inflammatory processes have only recently been recognized and evolving. In the context of parasitic diseases, PLTs function in directly responding to T. cruzi infection, and PLT interactions with immune cells in shaping the proinflammatory or immunoregulatory function of monocytes, macrophages, and neutrophils remains elusive. How T. cruzi infection alters systemic microenvironment conditions to influence PLT and immune-cell interactions is not understood. In this review, we discuss the current literature, and extrapolate the mechanistic situations to explain how PLT and innate immune cell (especially monocytes and macrophages) interactions might be sustaining hypercoagulability and thromboinflammation in chronic CD.
Collapse
Affiliation(s)
- Subhadip Choudhuri
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Nisha J Garg
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
12
|
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: 1.0] [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
|
13
|
Complex and Controversial Roles of Eicosanoids in Fungal Pathogenesis. J Fungi (Basel) 2021; 7:jof7040254. [PMID: 33800694 PMCID: PMC8065571 DOI: 10.3390/jof7040254] [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] [Received: 03/02/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/02/2023] Open
Abstract
The prevalence of fungal infections has increased in immunocompromised patients, leading to millions of deaths annually. Arachidonic acid (AA) metabolites, such as eicosanoids, play important roles in regulating innate and adaptative immune function, particularly since they can function as virulence factors enhancing fungal colonization and are produced by mammalian and lower eukaryotes, such as yeasts and other fungi (Candida albicans, Histoplasma capsulatum and Cryptococcus neoformans). C. albicans produces prostaglandins (PG), Leukotrienes (LT) and Resolvins (Rvs), whereas the first two have been well documented in Cryptococcus sp. and H. capsulatum. In this review, we cover the eicosanoids produced by the host and fungi during fungal infections. These fungal-derived PGs have immunomodulatory functions analogous to their mammalian counterparts. Prostaglandin E2 (PGE2) protects C. albicans and C. parapsilosis cells from the phagocytic and killing activity of macrophages. H. capsulatum PGs augment the fungal burden and host mortality rates in histoplasmosis. However, PGD2 potentiates the effects and production of LTB4, which is a very potent neutrophil chemoattractant that enhances host responses. Altogether, these data suggest that eicosanoids, mainly PGE2, may serve as a new potential target to combat diverse fungal infections.
Collapse
|
14
|
Tavares VDS, de Castro MV, Souza RDSO, Gonçalves IKA, Lima JB, Borges VDM, Araújo-Santos T. Lipid droplets of protozoan parasites: survival and pathogenicity. Mem Inst Oswaldo Cruz 2021; 116:e210270. [PMID: 35195194 PMCID: PMC8851939 DOI: 10.1590/0074-02760210270] [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: 08/11/2021] [Accepted: 12/14/2021] [Indexed: 12/04/2022] Open
Abstract
Lipid droplets (LDs; lipid bodies) are intracellular sites of lipid storage and metabolism present in all cell types. Eukaryotic LDs are involved in eicosanoid production during several inflammatory conditions, including infection by protozoan parasites. In parasites, LDs play a role in the acquisition of cholesterol and other neutral lipids from the host. The number of LDs increases during parasite differentiation, and the biogenesis of these organelles use specific signaling pathways involving protein kinases. In addition, LDs are important in cellular protection against lipotoxicity. Recently, these organelles have been implicated in eicosanoid and specialised lipid metabolism. In this article, we revise the main functions of protozoan parasite LDs and discuss future directions in the comprehension of these organelles in the context of pathogen virulence.
Collapse
Affiliation(s)
| | | | | | | | - Jonilson Berlink Lima
- Universidade Federal do Oeste da Bahia, Brasil; Fundação Oswaldo Cruz-Fiocruz, Brasil
| | | | - Théo Araújo-Santos
- Universidade Federal do Oeste da Bahia, Brasil; Fundação Oswaldo Cruz-Fiocruz, Brasil
| |
Collapse
|
15
|
Varikuti S, Jha BK, Holcomb EA, McDaniel JC, Karpurapu M, Srivastava N, McGwire BS, Satoskar AR, Parinandi NL. The role of vascular endothelium and exosomes in human protozoan parasitic diseases. ACTA ACUST UNITED AC 2020; 4. [PMID: 33089078 PMCID: PMC7575144 DOI: 10.20517/2574-1209.2020.27] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The vascular endothelium is a vital component in maintaining the structure and function of blood vessels. The endothelial cells (ECs) mediate vital regulatory functions such as the proliferation of cells, permeability of various tissue membranes, and exchange of gases, thrombolysis, blood flow, and homeostasis. The vascular endothelium also regulates inflammation and immune cell trafficking, and ECs serve as a replicative niche for many bacterial, viral, and protozoan infectious diseases. Endothelial dysfunction can lead to vasodilation and pro-inflammation, which are the hallmarks of many severe diseases. Exosomes are nanoscale membrane-bound vesicles that emerge from cells and serve as important extracellular components, which facilitate communication between cells and maintain homeostasis during normal and pathophysiological states. Exosomes are also involved in gene transfer, inflammation and antigen presentation, and mediation of the immune response during pathogenic states. Protozoa are a diverse group of unicellular organisms that cause many infectious diseases in humans. In this regard, it is becoming increasingly evident that many protozoan parasites (such as Plasmodium, Trypanosoma, Leishmania, and Toxoplasma) utilize exosomes for the transfer of their virulence factors and effector molecules into the host cells, which manipulate the host gene expression, immune responses, and other biological activities to establish and modulate infection. In this review, we discuss the role of the vascular endothelium and exosomes in and their contribution to pathogenesis in malaria, African sleeping sickness, Chagas disease, and leishmaniasis and toxoplasmosis with an emphasis on their actions on the innate and adaptive immune mechanisms of resistance.
Collapse
Affiliation(s)
- Sanjay Varikuti
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA.,Department of Bioscience & Biotechnology, Banasthali University, Banasthali 304022, India
| | - Bijay Kumar Jha
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Erin A Holcomb
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Jodi C McDaniel
- College of Nursing, The Ohio State University, Columbus, OH 43201, USA
| | - Manjula Karpurapu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Nidhi Srivastava
- Department of Bioscience & Biotechnology, Banasthali University, Banasthali 304022, India
| | - Bradford S McGwire
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH 43201, USA
| | - Narasimham L Parinandi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH 43201, USA
| |
Collapse
|
16
|
Booth LA, Smith TK. Lipid metabolism in Trypanosoma cruzi: A review. Mol Biochem Parasitol 2020; 240:111324. [PMID: 32961207 DOI: 10.1016/j.molbiopara.2020.111324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 01/08/2023]
Abstract
The cellular membranes of Trypanosoma cruzi, like all eukaryotes, contain varying amounts of phospholipids, sphingolipids, neutral lipids and sterols. A multitude of pathways exist for the de novo synthesis of these lipid families but Trypanosoma cruzi has also become adapted to scavenge some of these lipids from the host. Completion of the TriTryp genomes has led to the identification of many putative genes involved in lipid synthesis, revealing some interesting differences to higher eukaryotes. Although many enzymes involved in lipid synthesis have yet to be characterised, completed experiments have shown the indispensability of some lipid metabolic pathways. Furthermore, the bioactive lipids of Trypanosoma cruzi and their effects on the host are becoming increasingly studied. Further studies on lipid metabolism in Trypanosoma cruzi will no doubt reveal some attractive targets for therapeutic intervention as well as reveal the interplay between parasite lipids, host response and pathogenesis.
Collapse
Affiliation(s)
- Leigh-Ann Booth
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Scotland, KY16 9ST, United Kingdom
| | - Terry K Smith
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, Scotland, KY16 9ST, United Kingdom.
| |
Collapse
|
17
|
Niu M, Keller NP. Co-opting oxylipin signals in microbial disease. Cell Microbiol 2020; 21:e13025. [PMID: 30866138 DOI: 10.1111/cmi.13025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/01/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022]
Abstract
Oxylipins, or oxygenated lipids, are universal signalling molecules across all kingdoms of life. These molecules, either produced by microbial pathogens or their mammalian host, regulate inflammation during microbial infection. In this review, we summarise current literature on the biosynthesis pathways of microbial oxylipins and their biological activity towards mammalian cells. Collectively, these studies have illustrated how microbial pathogens can modulate immune rsponse and disease outcome via oxylipin-mediated mechanisms.
Collapse
Affiliation(s)
- Mengyao Niu
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Nancy P Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
18
|
Resolvin D1 Administration Is Beneficial in Trypanosoma cruzi Infection. Infect Immun 2020; 88:IAI.00052-20. [PMID: 32152197 DOI: 10.1128/iai.00052-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 03/02/2020] [Indexed: 12/14/2022] Open
Abstract
Chagas disease is a major public health issue, affecting ∼10 million people worldwide. Transmitted by a protozoan named Trypanosoma cruzi, this infection triggers a chronic inflammatory process that can lead to cardiomyopathy (Chagas disease). Resolvin D1 (RvD1) is a novel proresolution lipid mediator whose effects on inflammatory diseases dampens pathological inflammatory responses and can restore tissue homeostasis. Current therapies are not effective in altering the outcome of T. cruzi infection, and as RvD1 has been evaluated as a therapeutic agent in various inflammatory diseases, we examined if exogenous RvD1 could modulate the pathogenesis of Chagas disease in a murine model. CD-1 mice infected with the T. cruzi Brazil strain were treated with RvD1. Mice were administered 3 μg/kg of body weight RvD1 intraperitoneally on days 5, 10, and 15 to examine the effect of RvD1 on acute disease or administered the same dose on days 60, 65, and 70 to examine its effects on chronic infection. RvD1 therapy increased the survival rate and controlled parasite replication in mice with acute infection and reduced the levels of interferon gamma and transforming growth factor β (TGF-β) in mice with chronic infection. In addition, there was an increase in interleukin-10 levels with RvD1 therapy in both mice with acute infection and mice with chronic infection and a decrease in TGF-β levels and collagen content in cardiac tissue. Together, these data indicate that RvD1 therapy can dampen the inflammatory response, promote the resolution of T. cruzi infection, and prevent cardiac fibrosis.
Collapse
|
19
|
Abstract
Infectious myocarditis is the result of an immune response to a microbial infection of the heart. The blood vessels of the heart, both the intramyocardial microvasculature and the large epicardial coronary arteries, play an important role in the pathogenesis of infectious myocarditis. First of all, in addition to cardiomyocytes, endothelial cells of the cardiac (micro)vasculature are direct targets for infection. Moreover, through the expression of adhesion molecules and antigen presenting Major Histocompatibility Complex molecules, the blood vessels assist in shaping the cellular immune response in infectious myocarditis. In addition, damage and dysfunction of the cardiac (micro)vasculature are associated with thrombus formation as well as aberrant regulation of vascular tone including coronary vasospasm. These in turn can cause cardiac perfusion abnormalities and even myocardial infarction. In this review, we will discuss the role of the cardiac (micro)vasculature in the pathogenesis of infectious myocarditis.
Collapse
|
20
|
Varikuti S, Jha BK, Volpedo G, Ryan NM, Halsey G, Hamza OM, McGwire BS, Satoskar AR. Host-Directed Drug Therapies for Neglected Tropical Diseases Caused by Protozoan Parasites. Front Microbiol 2018; 9:2655. [PMID: 30555425 PMCID: PMC6284052 DOI: 10.3389/fmicb.2018.02655] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
The neglected tropical diseases (NTDs) caused by protozoan parasites are responsible for significant morbidity and mortality worldwide. Current treatments using anti-parasitic drugs are toxic and prolonged with poor patient compliance. In addition, emergence of drug-resistant parasites is increasing worldwide. Hence, there is a need for safer and better therapeutics for these infections. Host-directed therapy using drugs that target host pathways required for pathogen survival or its clearance is a promising approach for treating infections. This review will give a summary of the current status and advances of host-targeted therapies for treating NTDs caused by protozoa.
Collapse
Affiliation(s)
- Sanjay Varikuti
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Bijay Kumar Jha
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Greta Volpedo
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Nathan M Ryan
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Gregory Halsey
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Omar M Hamza
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Bradford S McGwire
- Division of Infectious Diseases, Department of Internal Medicine, The Ohio State University, Columbus, OH, United States
| | - Abhay R Satoskar
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Department of Microbiology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
21
|
Abstract
Endothelins were discovered more than thirty years ago as potent vasoactive compounds. Beyond their well-documented cardiovascular properties, however, the contributions of the endothelin pathway have been demonstrated in several neuroinflammatory processes and the peptides have been reported as clinically relevant biomarkers in neurodegenerative diseases. Several studies report that endothelin-1 significantly contributes to the progression of neuroinflammatory processes, particularly during infections in the central nervous system (CNS), and is associated with a loss of endothelial integrity at the blood brain barrier level. Because of the paucity of clinical trials with endothelin-1 antagonists in several infectious and non-infectious neuroinflammatory diseases, it remains an open question whether the 21 amino acid peptide is a mediator/modulator rather than a biomarker of the progression of neurodegeneration. This review focuses on the potential roles of endothelins in the pathology of neuroinflammatory processes, including infectious diseases of viral, bacterial or parasitic origin in which the synthesis of endothelins or its pharmacology have been investigated from the cell to the bedside in several cases, as well as in non-infectious inflammatory processes such as neurodegenerative disorders like Alzheimers Disease or central nervous system vasculitis.
Collapse
|
22
|
A role for trypanosomatid aldo-keto reductases in methylglyoxal, prostaglandin and isoprostane metabolism. Biochem J 2018; 475:2593-2610. [PMID: 30045874 PMCID: PMC6117947 DOI: 10.1042/bcj20180232] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 11/17/2022]
Abstract
Trypanosomatid parasites are the infectious agents causing Chagas disease, visceral and cutaneous leishmaniasis and human African trypanosomiasis. Recent work of others has implicated an aldo-keto reductase (AKR) in the susceptibility and resistance of Trypanosoma cruzi to benznidazole, a drug used to treat Chagas disease. Here, we show that TcAKR and homologues in the related parasites Trypanosoma brucei and Leishmania donovani do not reductively activate monocyclic (benznidazole, nifurtimox and fexinidazole) or bicyclic nitro-drugs such as PA-824. Rather, these enzymes metabolise a variety of toxic ketoaldehydes, such as glyoxal and methylglyoxal, suggesting a role in cellular defence against chemical stress. UPLC-QToF/MS analysis of benznidazole bioactivation by T. cruzi cell lysates confirms previous reports identifying numerous drug metabolites, including a dihydro-dihydroxy intermediate that can dissociate to form N-benzyl-2-guanidinoacetamide and glyoxal, a toxic DNA-glycating and cross-linking agent. Thus, we propose that TcAKR contributes to benznidazole resistance by the removal of toxic glyoxal. In addition, three of the four enzymes studied here display activity as prostaglandin F2α synthases, despite the fact that there are no credible cyclooxygenases in these parasites to account for formation of the precursor PGH2 from arachidonic acid. Our studies suggest that arachidonic acid is first converted non-enzymatically in parasite lysates to (PGH2-like) regioisomers by free radical-mediated peroxidation and that AKRs convert these lipid peroxides into isoprostanes, including prostaglandin F2α and 8-iso-prostaglandin F2α.
Collapse
|
23
|
López-Muñoz RA, Molina-Berríos A, Campos-Estrada C, Abarca-Sanhueza P, Urrutia-Llancaqueo L, Peña-Espinoza M, Maya JD. Inflammatory and Pro-resolving Lipids in Trypanosomatid Infections: A Key to Understanding Parasite Control. Front Microbiol 2018; 9:1961. [PMID: 30186271 PMCID: PMC6113562 DOI: 10.3389/fmicb.2018.01961] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/02/2018] [Indexed: 12/30/2022] Open
Abstract
Pathogenic trypanosomatids (Trypanosoma cruzi, Trypanosoma brucei, and Leishmania spp.) are protozoan parasites that cause neglected diseases affecting millions of people in Africa, Asia, and the Americas. In the process of infection, trypanosomatids evade and survive the immune system attack, which can lead to a chronic inflammatory state that induces cumulative damage, often killing the host in the long term. The immune mediators involved in this process are not entirely understood. Most of the research on the immunologic control of protozoan infections has been focused on acute inflammation. Nevertheless, when this process is not terminated adequately, permanent damage to the inflamed tissue may ensue. Recently, a second process, called resolution of inflammation, has been proposed to be a pivotal process in the control of parasite burden and establishment of chronic infection. Resolution of inflammation is an active process that promotes the normal function of injured or infected tissues. Several mediators are involved in this process, including eicosanoid-derived lipids, cytokines such as transforming growth factor (TGF)-β and interleukin (IL)-10, and other proteins such as Annexin-V. For example, during T. cruzi infection, pro-resolving lipids such as 15-epi-lipoxin-A4 and Resolvin D1 have been associated with a decrease in the inflammatory changes observed in experimental chronic heart disease, reducing inflammation and fibrosis, and increasing host survival. Furthermore, Resolvin D1 modulates the immune response in cells of patients with Chagas disease. In Leishmania spp. infections, pro-resolving mediators such as Annexin-V, lipoxins, and Resolvin D1 are related to the modulation of cutaneous manifestation of the disease. However, these mediators seem to have different roles in visceral or cutaneous leishmaniasis. Finally, although T. brucei infections are less well studied in terms of their relationship with inflammation, it has been found that arachidonic acid-derived lipids act as key regulators of the host immune response and parasite burden. Also, cytokines such as IL-10 and TGF-β may be related to increased infection. Knowledge about the inflammation resolution process is necessary to understand the host–parasite interplay, but it also offers an interesting opportunity to improve the current therapies, aiming to reduce the detrimental state induced by chronic protozoan infections.
Collapse
Affiliation(s)
- Rodrigo A López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Alfredo Molina-Berríos
- Instituto de Investigación en Ciencias Odontológicas, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Carolina Campos-Estrada
- Escuela de Química y Farmacia, Facultad de Farmacia, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigación Farmacopea Chilena, Universidad de Valparaíso, Valparaíso, Chile
| | - Patricio Abarca-Sanhueza
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Luis Urrutia-Llancaqueo
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Miguel Peña-Espinoza
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Juan D Maya
- Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
24
|
Lovo-Martins MI, Malvezi AD, Zanluqui NG, Lucchetti BFC, Tatakihara VLH, Mörking PA, de Oliveira AG, Goldenberg S, Wowk PF, Pinge-Filho P. Extracellular Vesicles Shed By Trypanosoma cruzi Potentiate Infection and Elicit Lipid Body Formation and PGE 2 Production in Murine Macrophages. Front Immunol 2018; 9:896. [PMID: 29755471 PMCID: PMC5934475 DOI: 10.3389/fimmu.2018.00896] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/11/2018] [Indexed: 12/16/2022] Open
Abstract
During the onset of Trypanosoma cruzi infection, an effective immune response is necessary to control parasite replication and ensure host survival. Macrophages have a central role in innate immunity, acting as an important trypanocidal cell and triggering the adaptive immune response through antigen presentation and cytokine production. However, T. cruzi displays immune evasion mechanisms that allow infection and replication in macrophages, favoring its chronic persistence. One potential mechanism is the release of T. cruzi strain Y extracellular vesicle (EV Y), which participate in intracellular communication by carrying functional molecules that signal host cells and can modulate the immune response. The present work aimed to evaluate immune modulation by EV Y in C57BL/6 mice, a prototype resistant to infection by T. cruzi strain Y, and the effects of direct EV Y stimulation of macrophages in vitro. EV Y inoculation in mice prior to T. cruzi infection resulted in increased parasitemia, elevated cardiac parasitism, decreased plasma nitric oxide (NO), reduced NO production by spleen cells, and modulation of cytokine production, with a reduction in TNF-α in plasma and decreased production of TNF-α and IL-6 by spleen cells from infected animals. In vitro assays using bone marrow-derived macrophages showed that stimulation with EV Y prior to infection by T. cruzi increased the parasite internalization rate and release of infective trypomastigotes by these cells. In this same scenario, EV Y induced lipid body formation and prostaglandin E2 (PGE2) production by macrophages even in the absence of T. cruzi. In infected macrophages, EV Y decreased production of PGE2 and cytokines TNF-α and IL-6 24 h after infection. These results suggest that EV Y modulates the host response in favor of the parasite and indicates a role for lipid bodies and PGE2 in immune modulation exerted by EVs.
Collapse
Affiliation(s)
- Maria Isabel Lovo-Martins
- Instituto Carlos Chagas, Fiocruz - Paraná, Curitiba, Brazil.,Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Aparecida Donizette Malvezi
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Bruno Fernando Cruz Lucchetti
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | - Vera Lúcia Hideko Tatakihara
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Admilton Gonçalves de Oliveira
- Laboratório de Microscopia Eletrônica e Microanálises, Central de Laboratórios de Pesquisa Multiusuários, Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Pryscilla Fanini Wowk
- Instituto Carlos Chagas, Fiocruz - Paraná, Curitiba, Brazil.,Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fiocruz - Paraná, Curitiba, Brazil
| | - Phileno Pinge-Filho
- Laboratório de Imunopatologia Experimental, Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina, Brazil
| |
Collapse
|
25
|
Lemos de Oliveira LF, Thackeray JT, Marin Neto JA, Dias Romano MM, Vieira de Carvalho EE, Mejia J, Tanaka DM, Kelly da Silva G, Abdalla DR, Malamut C, Bengel FM, de Lourdes Higuchi M, Schmidt A, Cunha-Neto E, Simões MV. Regional Myocardial Perfusion Disturbance in Experimental Chronic Chagas Cardiomyopathy. J Nucl Med 2018; 59:1430-1436. [DOI: 10.2967/jnumed.117.205450] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/22/2018] [Indexed: 11/16/2022] Open
|
26
|
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: 1.7] [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
|
27
|
Díaz-Viraqué F, Chiribao ML, Trochine A, González-Herrera F, Castillo C, Liempi A, Kemmerling U, Maya JD, Robello C. Old Yellow Enzyme from Trypanosoma cruzi Exhibits In Vivo Prostaglandin F 2α Synthase Activity and Has a Key Role in Parasite Infection and Drug Susceptibility. Front Immunol 2018; 9:456. [PMID: 29563916 PMCID: PMC5845897 DOI: 10.3389/fimmu.2018.00456] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/20/2018] [Indexed: 01/26/2023] Open
Abstract
The discovery that trypanosomatids, unicellular organisms of the order Kinetoplastida, are capable of synthesizing prostaglandins raised questions about the role of these molecules during parasitic infections. Multiple studies indicate that prostaglandins could be related to the infection processes and pathogenesis in trypanosomatids. This work aimed to unveil the role of the prostaglandin F2α synthase TcOYE in the establishment of Trypanosoma cruzi infection, the causative agent of Chagas disease. This chronic disease affects several million people in Latin America causing high morbidity and mortality. Here, we propose a prokaryotic evolutionary origin for TcOYE, and then we used in vitro and in vivo experiments to show that T. cruzi prostaglandin F2α synthase plays an important role in modulating the infection process. TcOYE overexpressing parasites were less able to complete the infective cycle in cell culture infections and increased cardiac tissue parasitic load in infected mice. Additionally, parasites overexpressing the enzyme increased PGF2α synthesis from arachidonic acid. Finally, an increase in benznidazole and nifurtimox susceptibility in TcOYE overexpressing parasites showed its participation in activating the currently anti-chagasic drugs, which added to its observed ability to confer resistance to hydrogen peroxide, highlights the relevance of this enzyme in multiple events including host-parasite interaction.
Collapse
Affiliation(s)
| | - María Laura Chiribao
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departamento de Bioquímica, Facultad de Medicina Universidad de la República, Montevideo, Uruguay
| | - Andrea Trochine
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Fabiola González-Herrera
- Programa de Farmacología Molecular y Clínica - ICBM, Facultad de Medicina Universidad de Chile, Santiago de Chile, Chile
| | - Christian Castillo
- Programa de Anatomía y Biología del Desarrollo - ICBM, Facultad de Medicina Universidad De Chile, Santiago de Chile, Chile
| | - Ana Liempi
- Programa de Anatomía y Biología del Desarrollo - ICBM, Facultad de Medicina Universidad De Chile, Santiago de Chile, Chile
| | - Ulrike Kemmerling
- Programa de Anatomía y Biología del Desarrollo - ICBM, Facultad de Medicina Universidad De Chile, Santiago de Chile, Chile
| | - Juan Diego Maya
- Programa de Farmacología Molecular y Clínica - ICBM, Facultad de Medicina Universidad de Chile, Santiago de Chile, Chile
| | - Carlos Robello
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departamento de Bioquímica, Facultad de Medicina Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
28
|
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
|
29
|
Protective effect of aspirin treatment on mouse behavior in the acute phase of experimental infection with Trypanosoma cruzi. Parasitol Res 2017; 117:189-200. [PMID: 29196837 DOI: 10.1007/s00436-017-5693-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 11/21/2017] [Indexed: 12/16/2022]
Abstract
Chagas disease is a potentially fatal disease caused by the parasite Trypanosoma cruzi, which can in some cases affect the central nervous system. The objective was to evaluate the effect of aspirin (ASA) in the behavior of mice infected with T. cruzi during the acute phase. This was an experimental study with random assignation. Twenty four BALB/c mice were divided into four groups of six animals each as follows: only ASA (OA), ASA before infection (BI), ASA after infection (AI) and only infection (OI). The strain used for infection was M/HOM/Bra/53/Y. An ASA dose of 100 mg/kg per day was administered 72 h before infection to BI group and the same dose 48 h after infection to AI group. Mice behavior in the open field test, mortality, and brain histopathology was evaluated. Data were analyzed using ANOVA, chi square test, and Kaplan-Meier with long-rank for survival analysis. In the open field test, the OA group has similar results with the BI group, in the variables of immobility and escape. Also, the OA group displayed significantly higher rates of micturition (p < 0.001) and defecation (p < 0.001) compared to infected groups. Mortality was higher in BI group (p = 0.02). The presence of T. cruzi amastigotes were higher in brain tissues of the AI and OI groups (p = 0.008). In conclusion, the administration of ASA before infection seemed to prevent behavioral changes induced by the acute infection, but it led to accelerated mortality. The study highlighted the potential importance of the pathways inhibited by ASA in the early hours of acute infection with T. cruzi.
Collapse
|
30
|
Biagiotti M, Dominguez S, Yamout N, Zufferey R. Lipidomics and anti-trypanosomatid chemotherapy. Clin Transl Med 2017; 6:27. [PMID: 28766182 PMCID: PMC5539062 DOI: 10.1186/s40169-017-0160-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/26/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Trypanosomatids such as Leishmania, Trypanosoma brucei and Trypanosoma cruzi belong to the order Kinetoplastida and are the source of many significant human and animal diseases. Current treatment is unsatisfactory and is compromised by the rising appearance of drug resistant parasites. Novel and more effective chemotherapeutics are urgently needed to treat and prevent these devastating diseases, which relies on the identification of essential, parasite specific targets that are absent in the host. Lipids constitute essential components of the cell and carry out multiple critical functions from building blocks of biological membranes to regulatory roles in signal transduction, organellar biogenesis, energy storage, and virulence. The recent technological advances of lipidomics has facilitated the broadening of our knowledge in the field of cellular lipid content, structure, functions, and metabolic pathways. MAIN BODY This review highlights the application of lipidomics (i) in the characterization of the lipidome of kinetoplastid parasites or of their subcellular structure(s), (ii) in the identification of unique lipid species or metabolic pathways that can be targeted for novel drug therapies, (iii) as an analytic tool to gain a deeper insight into the roles of specific enzymes in lipid metabolism using genetically modified microorganisms, and (iv) in deciphering the mechanism of action of anti-microbial drugs on lipid metabolism. Lastly, an outlook stating where the field is evolving is presented. CONCLUSION Lipidomics has contributed to the expanding knowledge related to lipid metabolism, mechanism of drug action and resistance, and pathogen-host interaction of trypanosomatids, which provides a solid basis for the development of better anti-parasitic pharmaceuticals.
Collapse
Affiliation(s)
| | | | - Nader Yamout
- St John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Rachel Zufferey
- St John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| |
Collapse
|
31
|
Silva JF, Capettini LSA, da Silva JFP, Sales-Junior P, Cruz JS, Cortes SF, Lemos VS. Mechanisms of vascular dysfunction in acute phase of Trypanosoma cruzi infection in mice. Vascul Pharmacol 2016; 82:73-81. [PMID: 26988253 DOI: 10.1016/j.vph.2016.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/27/2015] [Accepted: 03/08/2016] [Indexed: 12/12/2022]
Abstract
Vascular disorders have a direct link to mortality in the acute phase of Trypanosoma cruzi infection. However, the underlying mechanisms of vascular dysfunction in this phase are largely unknown. We hypothesize that T. cruzi invades endothelial cells causing dysfunction in contractility and relaxation of the mouse aorta. Immunodetection of T. cruzi antigen TcRBP28 was observed in endothelial cells. There was a decreased endothelial nitric oxide synthase (eNOS)-derived NO-dependent vascular relaxation, and increased vascular contractility accompanied by augmented superoxide anions production. Endothelial removal, inhibition of cyclooxygenase 2 (COX-2), blockade of thromboxane A2 (TXA2) TP receptors, and scavenger of superoxide normalized the contractile response. COX-2, thromboxane synthase, inducible nitric oxide synthase (iNOS), p65 NFκB subunit and p22(phox) of NAD(P)H oxidase (NOX) subunit expressions were increased in vessels of chagasic animals. Serum TNF-α was augmented. Basal NO production, and nitrotyrosine residue expression were increased. It is concluded that T. cruzi invades mice aorta endothelial cells and increases TXA2/TP receptor/NOX-derived superoxide formation. Alongside, T. cruzi promotes systemic TNF-α increase, which stimulates iNOS expression in vessels and nitrosative stress. In light of the heart failure that develops in the chronic phase of the disease, to understand the mechanism involved in the increased contractility of the aorta is crucial.
Collapse
Affiliation(s)
- Josiane F Silva
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Luciano S A Capettini
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil; Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - José F P da Silva
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | | | - Jader Santos Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Steyner F Cortes
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Virginia S Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil.
| |
Collapse
|
32
|
Bonney KM, Engman DM. Autoimmune pathogenesis of Chagas heart disease: looking back, looking ahead. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1537-47. [PMID: 25857229 DOI: 10.1016/j.ajpath.2014.12.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 12/21/2014] [Accepted: 12/23/2014] [Indexed: 01/14/2023]
Abstract
Chagas heart disease is an inflammatory cardiomyopathy that develops in approximately one-third of individuals infected with the protozoan parasite Trypanosoma cruzi. Since the discovery of T. cruzi by Carlos Chagas >100 years ago, much has been learned about Chagas disease pathogenesis; however, the outcome of T. cruzi infection is highly variable and difficult to predict. Many mechanisms have been proposed to promote tissue inflammation, but the determinants and the relative importance of each have yet to be fully elucidated. The notion that some factor other than the parasite significantly contributes to the development of myocarditis was hypothesized by the first physician-scientists who noted the conspicuous absence of parasites in the hearts of those who succumbed to Chagas disease. One of these factors-autoimmunity-has been extensively studied for more than half a century. Although questions regarding the functional role of autoimmunity in the pathogenesis of Chagas disease remain unanswered, the development of autoimmune responses during infection clearly occurs in some individuals, and the implications that this autoimmunity may be pathogenic are significant. In this review, we summarize what is known about the pathogenesis of Chagas heart disease and conclude with a view of the future of Chagas disease diagnosis, pathogenesis, therapy, and prevention, emphasizing recent advances in these areas that aid in the management of Chagas disease.
Collapse
Affiliation(s)
- Kevin M Bonney
- Department of Pathology, Northwestern University, Chicago, Illinois; Department of Microbiology-Immunology, Northwestern University, Chicago, Illinois
| | - David M Engman
- Department of Pathology, Northwestern University, Chicago, Illinois; Department of Microbiology-Immunology, Northwestern University, Chicago, Illinois; Department of Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois.
| |
Collapse
|
33
|
Moraes KCM, Diniz LF, Bahia MT. Role of cyclooxygenase-2 in Trypanosoma cruzi survival in the early stages of parasite host-cell interaction. Mem Inst Oswaldo Cruz 2015; 110:181-91. [PMID: 25946241 PMCID: PMC4489448 DOI: 10.1590/0074-02760140311] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 03/18/2015] [Indexed: 12/22/2022] Open
Abstract
Chagas disease, caused by the intracellular protozoan Trypanosoma cruzi, is a serious health problem in Latin America. During this parasitic infection, the heart is one of the major organs affected. The pathogenesis of tissue remodelling, particularly regarding cardiomyocyte behaviour after parasite infection and the molecular mechanisms that occur immediately following parasite entry into host cells are not yet completely understood. When cells are infected with T. cruzi, they develop an inflammatory response, in which cyclooxygenase-2 (COX-2) catalyses rate-limiting steps in the arachidonic acid pathway. However, how the parasite interaction modulates COX-2 activity is poorly understood. In this study, the H9c2 cell line was used as our model and we investigated cellular and biochemical aspects during the initial 48 h of parasitic infection. Oscillatory activity of COX-2 was observed, which correlated with the control of the pro-inflammatory environment in infected cells. Interestingly, subcellular trafficking was also verified, correlated with the control of Cox-2 mRNA or the activated COX-2 protein in cells, which is directly connected with the assemble of stress granules structures. Our collective findings suggest that in the very early stage of the T. cruzi-host cell interaction, the parasite is able to modulate the cellular metabolism in order to survives.
Collapse
Affiliation(s)
- Karen CM Moraes
- Laboratório de Biologia Molecular, Departamento de Biologia, Instituto
de Biociências, Universidade Estadual Paulista Júlio de Mesquita Filho, Rio Claro, SP,
Brasil
| | - Lívia F Diniz
- Laboratório de Doença de Chagas, Departamento de Ciências Biológicas,
Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro
Preto, MG, Brasil
| | - Maria Terezinha Bahia
- Laboratório de Doença de Chagas, Departamento de Ciências Biológicas,
Núcleo de Pesquisa em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro
Preto, MG, Brasil
| |
Collapse
|
34
|
Gazos-Lopes F, Oliveira MM, Hoelz LVB, Vieira DP, Marques AF, Nakayasu ES, Gomes MT, Salloum NG, Pascutti PG, Souto-Padrón T, Monteiro RQ, Lopes AH, Almeida IC. Structural and functional analysis of a platelet-activating lysophosphatidylcholine of Trypanosoma cruzi. PLoS Negl Trop Dis 2014; 8:e3077. [PMID: 25101628 PMCID: PMC4125143 DOI: 10.1371/journal.pntd.0003077] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 06/13/2014] [Indexed: 12/15/2022] Open
Abstract
Background Trypanosoma cruzi is the causative agent of the life-threatening Chagas disease, in which increased platelet aggregation related to myocarditis is observed. Platelet-activating factor (PAF) is a potent intercellular lipid mediator and second messenger that exerts its activity through a PAF-specific receptor (PAFR). Previous data from our group suggested that T. cruzi synthesizes a phospholipid with PAF-like activity. The structure of T. cruzi PAF-like molecule, however, remains elusive. Methodology/Principal findings Here, we have purified and structurally characterized the putative T. cruzi PAF-like molecule by electrospray ionization-tandem mass spectrometry (ESI-MS/MS). Our ESI-MS/MS data demonstrated that the T. cruzi PAF-like molecule is actually a lysophosphatidylcholine (LPC), namely sn-1 C18:1(delta 9)-LPC. Similar to PAF, the platelet-aggregating activity of C18:1-LPC was abrogated by the PAFR antagonist, WEB 2086. Other major LPC species, i.e., C16:0-, C18:0-, and C18:2-LPC, were also characterized in all T. cruzi stages. These LPC species, however, failed to induce platelet aggregation. Quantification of T. cruzi LPC species by ESI-MS revealed that intracellular amastigote and trypomastigote forms have much higher levels of C18:1-LPC than epimastigote and metacyclic trypomastigote forms. C18:1-LPC was also found to be secreted by the parasite in extracellular vesicles (EV) and an EV-free fraction. A three-dimensional model of PAFR was constructed and a molecular docking study was performed to predict the interactions between the PAFR model and PAF, and each LPC species. Molecular docking data suggested that, contrary to other LPC species analyzed, C18:1-LPC is predicted to interact with the PAFR model in a fashion similar to PAF. Conclusions/Significance Taken together, our data indicate that T. cruzi synthesizes a bioactive C18:1-LPC, which aggregates platelets via PAFR. We propose that C18:1-LPC might be an important lipid mediator in the progression of Chagas disease and its biosynthesis could eventually be exploited as a potential target for new therapeutic interventions. Chagas disease, caused by the parasite Trypanosoma cruzi, was exclusively confined to Latin America but it has recently spread to other regions of the world. Chagas disease affects 8–10 million people and kills thousands of them every year. Lysophosphatidylcholine (LPC) is a major bioactive phospholipid of human plasma low-density lipoproteins (LDL). Platelet-activating factor (PAF) is a phospholipid similar to LPC and a potent intercellular mediator. Both PAF and LPC have been reported to act on mammalian cells through PAF receptor (PAFR). Previous data from our group suggested that T. cruzi produces a phospholipid with PAF activity. Here, we describe the structural and functional analysis of different species of LPC from T. cruzi, including a LPC with a fatty acid chain of 18 carbon atoms and one double bond (C18:1-LPC). We also show that C18:1-LPC is able to induce rabbit platelet aggregation, which is abrogated by a PAFR antagonist. In addition, a three-dimensional model of human PAFR was constructed. Contrary to other T. cruzi LPC molecules, C18:1-LPC is predicted to interact with the PAFR model in a fashion similar to PAF. Further studies are needed to validate the biosynthesis of T. cruzi C18:1-LPC as a potential drug target in Chagas disease.
Collapse
Affiliation(s)
- Felipe Gazos-Lopes
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
| | - Mauricio M. Oliveira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas V. B. Hoelz
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco G, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle P. Vieira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre F. Marques
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Parasitologia, Pampulha, Belo Horizonte, Minas Gerais, Brazil
| | - Ernesto S. Nakayasu
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
| | - Marta T. Gomes
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco H, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nasim G. Salloum
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
| | - Pedro G. Pascutti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco G, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thaïs Souto-Padrón
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Q. Monteiro
- Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco H, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Angela H. Lopes
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Cidade Universitária, Centro de Ciências da Saúde, Bloco I, Ilha do Fundão, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: (AHL); (ICA)
| | - Igor C. Almeida
- The Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso (UTEP), El Paso, Texas, United States of America
- * E-mail: (AHL); (ICA)
| |
Collapse
|
35
|
Nisimura LM, Estato V, de Souza EM, Reis PA, Lessa MA, Castro-Faria-Neto HC, Pereira MCDS, Tibiriçá E, Garzoni LR. Acute Chagas disease induces cerebral microvasculopathy in mice. PLoS Negl Trop Dis 2014; 8:e2998. [PMID: 25010691 PMCID: PMC4091872 DOI: 10.1371/journal.pntd.0002998] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 05/30/2014] [Indexed: 12/16/2022] Open
Abstract
Cardiomyopathy is the main clinical form of Chagas disease (CD); however, cerebral manifestations, such as meningoencephalitis, ischemic stroke and cognitive impairment, can also occur. The aim of the present study was to investigate functional microvascular alterations and oxidative stress in the brain of mice in acute CD. Acute CD was induced in Swiss Webster mice (SWM) with the Y strain of Trypanosoma cruzi (T. cruzi). Cerebral functional capillary density (the number of spontaneously perfused capillaries), leukocyte rolling and adhesion and the microvascular endothelial-dependent response were analyzed over a period of fifteen days using intravital video-microscopy. We also evaluated cerebral oxidative stress with the thiobarbituric acid reactive species TBARS method. Compared with the non-infected group, acute CD significantly induced cerebral functional microvascular alterations, including (i) functional capillary rarefaction, (ii) increased leukocyte rolling and adhesion, (iii) the formation of microvascular platelet-leukocyte aggregates, and (iv) alteration of the endothelial response to acetylcholine. Moreover, cerebral oxidative stress increased in infected animals. We concluded that acute CD in mice induced cerebral microvasculopathy, characterized by a reduced incidence of perfused capillaries, a high number of microvascular platelet-leukocyte aggregates, a marked increase in leukocyte-endothelium interactions and brain arteriolar endothelial dysfunction associated with oxidative stress. These results suggest the involvement of cerebral microcirculation alterations in the neurological manifestations of CD. Chagas disease (CD) is a neglected tropical illness caused by the parasite Trypanosoma cruzi (T. cruzi). It is endemic in Latin America and affects 10 million people worldwide. Meningoencephalitis occurs in children with acute CD and in immunosuppressed patients suffering acute CD reactivation. During the chronic phase, cerebral manifestations, including ischemic stroke and cognitive impairment, can also occur. Although microvascular alterations have been implicated in Chagas cardiomyopathy, the main clinical form of the disease, there is a lack of discussion in some studies regarding alterations of the cerebral microcirculation in CD. In the present study, we evaluated the functionality of the cerebral microcirculation in mice infected by T. cruzi. Utilizing an intravital video-microscope, we observed in the brain of infected mice a reduction in the number of perfused capillaries, an increased interaction between inflammatory cells and venules, the presence of microvascular platelet-leukocyte aggregates and alterations in the dilatation capacity of arterioles. Moreover, cerebral oxidative stress was increased in infected animals. We concluded that acute CD induced cerebral microvasculopathy.
Collapse
Affiliation(s)
- Lindice Mitie Nisimura
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Estato
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elen Mello de Souza
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia A. Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Adriano Lessa
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hugo Caire Castro-Faria-Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mirian Claudia de Souza Pereira
- Laboratório de Ultra-estrutura Celular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo Tibiriçá
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Ribeiro Garzoni
- Laboratório de Investigação Cardiovascular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail: ,
| |
Collapse
|
36
|
Aspirin modulates innate inflammatory response and inhibits the entry of Trypanosoma cruzi in mouse peritoneal macrophages. Mediators Inflamm 2014; 2014:580919. [PMID: 25045211 PMCID: PMC4089847 DOI: 10.1155/2014/580919] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/13/2014] [Accepted: 05/20/2014] [Indexed: 01/09/2023] Open
Abstract
The intracellular protozoan parasite Trypanosoma cruzi causes Chagas disease, a serious disorder that affects millions of people in Latin America. Cell invasion by T. cruzi and its intracellular replication are essential to the parasite's life cycle and for the development of Chagas disease. Here, we present evidence suggesting the involvement of the host's cyclooxygenase (COX) enzyme during T. cruzi invasion. Pharmacological antagonist for COX-1, aspirin (ASA), caused marked inhibition of T. cruzi infection when peritoneal macrophages were pretreated with ASA for 30 min at 37°C before inoculation. This inhibition was associated with increased production of IL-1β and nitric oxide (NO(∙)) by macrophages. The treatment of macrophages with either NOS inhibitors or prostaglandin E2 (PGE2) restored the invasive action of T. cruzi in macrophages previously treated with ASA. Lipoxin ALX-receptor antagonist Boc2 reversed the inhibitory effect of ASA on trypomastigote invasion. Our results indicate that PGE2, NO(∙), and lipoxins are involved in the regulation of anti-T. cruzi activity by macrophages, providing a better understanding of the role of prostaglandins in innate inflammatory response to T. cruzi infection as well as adding a new perspective to specific immune interventions.
Collapse
|
37
|
Araújo-Santos T, Rodríguez NE, Moura-Pontes S, Dixt UG, Abánades DR, Bozza PT, Wilson ME, Borges VM. Role of prostaglandin F2α production in lipid bodies from Leishmania infantum chagasi: insights on virulence. J Infect Dis 2014; 210:1951-61. [PMID: 24850789 DOI: 10.1093/infdis/jiu299] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Lipid bodies (LB; lipid droplets) are cytoplasmic organelles involved in lipid metabolism. Mammalian LBs display an important role in host-pathogen interactions, but the role of parasite LBs in biosynthesis of prostaglandin F2α (PGF2α) has not been investigated. We report herein that LBs increased in abundance during development of Leishmania infantum chagasi to a virulent metacyclic stage, as did the expression of PGF2α synthase (PGFS). The amount of parasite LBs and PGF2α were modulated by exogenous arachidonic acid. During macrophage infection, LBs were restricted to parasites inside the parasitophorous vacuoles (PV). We detected PGF2α receptor (FP) on the Leishmania PV surface. The blockage of FP with AL8810, a selective antagonist, hampered Leishmania infection, whereas the irreversible inhibition of cyclooxygenase with aspirin increased the parasite burden. These data demonstrate novel functions for parasite-derived LBs and PGF2α in the cellular metabolism of Leishmania and its evasion of the host immune response.
Collapse
Affiliation(s)
- Théo Araújo-Santos
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation (FIOCRUZ) Federal University of Bahia (UFBA), Salvador, Bahia, Brazil University of Iowa and the Iowa City VA Medical Center, Iowa
| | | | - Sara Moura-Pontes
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation (FIOCRUZ) Federal University of Bahia (UFBA), Salvador, Bahia, Brazil
| | | | - Daniel R Abánades
- Department of Chemical and Physical Biology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | | - Mary E Wilson
- University of Iowa and the Iowa City VA Medical Center, Iowa
| | - Valéria Matos Borges
- Gonçalo Moniz Research Center, Oswaldo Cruz Foundation (FIOCRUZ) Federal University of Bahia (UFBA), Salvador, Bahia, Brazil Institute for Investigation in Immunology, iii-INCT (National Institute of Science and Technology), São Paulo, Brazil
| |
Collapse
|
38
|
Endothelin-1 and its role in the pathogenesis of infectious diseases. Life Sci 2014; 118:110-9. [PMID: 24780317 DOI: 10.1016/j.lfs.2014.04.021] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/08/2014] [Accepted: 04/15/2014] [Indexed: 12/12/2022]
Abstract
Endothelins are potent regulators of vascular tone, which also have mitogenic, apoptotic, and immunomodulatory properties (Rubanyi and Polokoff, 1994; Kedzierski and Yanagisawa, 2001; Bagnato et al., 2011). Three isoforms of endothelin have been identified to date, with endothelin-1 (ET-1) being the best studied. ET-1 is classically considered a potent vasoconstrictor. However, in addition to the effects of ET-1 on vascular smooth muscle cells, the peptide is increasingly recognized as a pro-inflammatory cytokine (Teder and Noble, 2000; Sessa et al., 1991). ET-1 causes platelet aggregation and plays a role in the increased expression of leukocyte adhesion molecules, the synthesis of inflammatory mediators contributing to vascular dysfunction. High levels of ET-1 are found in alveolar macrophages, leukocytes (Sessa et al., 1991) and fibroblasts (Gu et al., 1991). Clinical and experimental data indicate that ET-1 is involved in the pathogenesis of sepsis (Tschaikowsky et al., 2000; Goto et al., 2012), viral and bacterial pneumonia (Schuetz et al., 2008; Samransamruajkit et al., 2002), Rickettsia conorii infections (Davi et al., 1995), Chagas disease (Petkova et al., 2000, 2001), and severe malaria (Dai et al., 2012; Machado et al., 2006; Wenisch et al., 1996a; Dietmann et al., 2008). In this minireview, we will discuss the role of endothelin in the pathogenesis of infectious processes.
Collapse
|
39
|
Requena-Méndez A, López MC, Angheben A, Izquierdo L, Ribeiro I, Pinazo MJ, Gascon J, Muñoz J. Evaluating Chagas disease progression and cure through blood-derived biomarkers: a systematic review. Expert Rev Anti Infect Ther 2014; 11:957-76. [DOI: 10.1586/14787210.2013.824718] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
40
|
Molina-Berríos A, Campos-Estrada C, Lapier M, Duaso J, Kemmerling U, Galanti N, Leiva M, Ferreira J, López-Muñoz R, Maya JD. Benznidazole prevents endothelial damage in an experimental model of Chagas disease. Acta Trop 2013; 127:6-13. [PMID: 23529066 DOI: 10.1016/j.actatropica.2013.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/06/2013] [Accepted: 03/15/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To evaluate the effect of benznidazole on endothelial activation in a murine model of Chagas disease. METHODS A low (30mg/kg/day) and a high (100mg/kg/day) dose of benznidazole were administered to mice infected with Trypanosoma cruzi during the early phases of the infection. The effects of the treatments were assessed at 24 and 90 days postinfection by evaluating the parasitaemia, mortality, histopathological changes and expression of ICAM in the cardiac tissue. The blood levels of thromboxane A2, soluble ICAM and E-selectin were also measured. T. cruzi clearance was assessed by the detection of parasite DNA in the heart tissue of infected mice. RESULTS Benznidazole decreased the cardiac damage induced by the parasite, and amastigote nests disappeared at 90 days postinfection. Both doses cleared the parasite from the cardiac tissue at 24 and 90 days postinfection. In addition, benznidazole decreased the thromboxane levels and normalized the plasma sICAM and sE-selectin levels by 90 days postinfection. CONCLUSIONS Early administration of benznidazole at a dose as low as 30mg/kg eradicates T. cruzi from cardiac tissue. Additionally, benznidazole prevents cardiac damage and modulates endothelial activation as part of its antichagasic activity.
Collapse
|
41
|
Rub A, Arish M, Husain SA, Ahmed N, Akhter Y. Host-lipidome as a potential target of protozoan parasites. Microbes Infect 2013; 15:649-60. [PMID: 23811020 DOI: 10.1016/j.micinf.2013.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/13/2013] [Accepted: 06/18/2013] [Indexed: 12/24/2022]
Abstract
Host-lipidome caters parasite interaction by acting as first line of recognition, attachment on the cell surface, intracellular trafficking, and survival of the parasite inside the host cell. Here, we summarize how protozoan parasites exploit host-lipidome by suppressing, augmenting, engulfing, remodeling and metabolizing lipids to achieve successful parasitism inside the host.
Collapse
Affiliation(s)
- Abdur Rub
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India.
| | | | | | | | | |
Collapse
|
42
|
Abstract
Cryptosporidium is a protozoan parasite of medical and veterinary importance that causes gastroenteritis in a variety of vertebrate hosts. Several studies have reported different degrees of pathogenicity and virulence among Cryptosporidium species and isolates of the same species as well as evidence of variation in host susceptibility to infection. The identification and validation of Cryptosporidium virulence factors have been hindered by the renowned difficulties pertaining to the in vitro culture and genetic manipulation of this parasite. Nevertheless, substantial progress has been made in identifying putative virulence factors for Cryptosporidium. This progress has been accelerated since the publication of the Cryptosporidium parvum and C. hominis genomes, with the characterization of over 25 putative virulence factors identified by using a variety of immunological and molecular techniques and which are proposed to be involved in aspects of host-pathogen interactions from adhesion and locomotion to invasion and proliferation. Progress has also been made in the contribution of host factors that are associated with variations in both the severity and risk of infection. Here we provide a review comprised of the current state of knowledge on Cryptosporidium infectivity, pathogenesis, and transmissibility in light of our contemporary understanding of microbial virulence.
Collapse
|
43
|
Molina-Berríos A, Campos-Estrada C, Lapier M, Duaso J, Kemmerling U, Galanti N, Ferreira J, Morello A, López-Muñoz R, Maya JD. Protection of vascular endothelium by aspirin in a murine model of chronic Chagas' disease. Parasitol Res 2013; 112:2731-9. [PMID: 23681190 DOI: 10.1007/s00436-013-3444-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 04/26/2013] [Indexed: 12/22/2022]
Abstract
Chronic Chagas' disease affects 10-30 % of patients infected with Trypanosoma cruzi, and it mainly manifests as cardiomyopathy. Important pathophysiological mechanisms involved in the cardiac lesions include activation of the endothelium and induced microvascular alterations. These processes involve the production of endothelial adhesion molecules and thromboxane A2, which are involved in inflammatory cell recruitment and platelet aggregation, respectively. Cyclooxygenase inhibitors such as aspirin decrease thromboxane production and alter the course of Chagas' disease, both in the acute and chronic phases. We studied the effects of the administration of low and high doses of aspirin during the early phase of T. cruzi infection, following microvascular damage in the context of a chronic murine model of Chagas' disease. The effects of both schedules were assessed at 24 and 90 days postinfection by evaluating parasitemia, mortality, and cardiac histopathological changes as well as the expression of ICAM, VCAM, and E-selectin in cardiac tissue. Thromboxane A2, soluble ICAM, and E-selectin blood levels were also measured. While aspirin did not affect parasitemia or mortality in the infected mice, it decreased both cardiac inflammatory infiltrates and thromboxane levels. Additionally, at 90 days postinfection, aspirin normalized sICAM and sE-selectin levels. Considering the improved endothelial function induced by aspirin, we propose the possibility of including this drug in clinical therapy to treat chronic Chagas' disease.
Collapse
Affiliation(s)
- Alfredo Molina-Berríos
- Molecular and Clinical Pharmacology Program, Biomedical Sciences Institute (ICBM), Faculty of Medicine, University of Chile, Independencia 1027, Santiago, Chile
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Molina-Berríos A, Campos-Estrada C, Henriquez N, Faúndez M, Torres G, Castillo C, Escanilla S, Kemmerling U, Morello A, López-Muñoz RA, Maya JD. Protective role of acetylsalicylic acid in experimental Trypanosoma cruzi infection: evidence of a 15-epi-lipoxin A₄-mediated effect. PLoS Negl Trop Dis 2013; 7:e2173. [PMID: 23638194 PMCID: PMC3630130 DOI: 10.1371/journal.pntd.0002173] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 03/08/2013] [Indexed: 01/03/2023] Open
Abstract
Chagas' disease, produced by Trypanosoma cruzi, affects more than 8 million people, producing approximately 10,000 deaths each year in Latin America. Migration of people from endemic regions to developed countries has expanded the risk of infection, transforming this disease into a globally emerging problem. PGE₂ and other eicosanoids contribute to cardiac functional deficits after infection with T. cruzi. Thus, the inhibition of host cyclooxygenase (COX) enzyme emerges as a potential therapeutic target. In vivo studies about the effect of acetylsalicylic acid (ASA) upon T. cruzi infection are controversial, and always report the effect of ASA at a single dose. Therefore, we aimed to analyze the effect of ASA at different doses in an in vivo model of infection and correlate it with the production of arachidonic acid metabolites. ASA decreased mortality, parasitemia, and heart damage in T. cruzi (Dm28c) infected mice, at the low doses of 25 and 50 mg/Kg. However, this effect disappeared when the high ASA doses of 75 and 100 mg/Kg were used. We explored whether this observation was related to the metabolic shift toward the production of 5-lipoxygenase derivatives, and although we did not observe an increase in LTB4 production in infected RAW cells and mice infected, we did find an increase in 15-epi-LXA₄ (an ASA-triggered lipoxin). We also found high levels of 15-epi-LXA₄ in T. cruzi infected mice treated with the low doses of ASA, while the high ASA doses decreased 15-epi-LXA₄ levels. Importantly, 15-epi-LXA₄ prevented parasitemia, mortality, and cardiac changes in vivo and restored the protective role in the treatment with a high dose of ASA. This is the first report showing the production of ASA-triggered lipoxins in T. cruzi infected mice, which demonstrates the role of this lipid as an anti-inflammatory molecule in the acute phase of the disease.
Collapse
Affiliation(s)
- Alfredo Molina-Berríos
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Diego Portales, Santiago, Chile
| | - Carolina Campos-Estrada
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Natalia Henriquez
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Mario Faúndez
- Departamento de Farmacia, Facultad de Química. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gloria Torres
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Christian Castillo
- Anatomy and Developmental Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sebastián Escanilla
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Ulrike Kemmerling
- Anatomy and Developmental Biology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Antonio Morello
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Rodrigo A. López-Muñoz
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan D. Maya
- Molecular and Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
45
|
The absence of myocardial calcium-independent phospholipase A2γ results in impaired prostaglandin E2 production and decreased survival in mice with acute Trypanosoma cruzi infection. Infect Immun 2013; 81:2278-87. [PMID: 23429536 DOI: 10.1128/iai.00497-12] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cardiomyopathy is a serious complication of Chagas' disease, caused by the protozoan parasite Trypanosoma cruzi. The parasite often infects cardiac myocytes, causing the release of inflammatory mediators, including eicosanoids. A recent study from our laboratory demonstrated that calcium-independent phospholipase A2γ (iPLA2γ) accounts for the majority of PLA2 activity in rabbit ventricular myocytes and is responsible for arachidonic acid (AA) and prostaglandin E2 (PGE2) release. Thus, we hypothesized that cardiac iPLA2γ contributes to eicosanoid production in T. cruzi infection. Inhibition of the isoform iPLA2γ or iPLA2β, with the R or S enantiomer of bromoenol lactone (BEL), respectively, demonstrated that iPLA2γ is the predominant isoform in immortalized mouse cardiac myocytes (HL-1 cells). Stimulation of HL-1 cells with thrombin, a serine protease associated with microthrombus formation in Chagas' disease and a known activator of iPLA2, increased AA and PGE2 release, accompanied by platelet-activating factor (PAF) production. Similarly, T. cruzi infection resulted in increased AA and PGE2 release over time that was inhibited by pretreatment with (R)-BEL. Further, T. cruzi-infected iPLA2γ-knockout (KO) mice had lower survival rates and increased tissue parasitism compared to wild-type (WT) mice, suggesting that iPLA2γ-KO mice were more susceptible to infection than WT mice. A significant increase in iPLA2 activity was observed in WT mice following infection, whereas iPLA2γ-KO mice showed no alteration in cardiac iPLA2 activity and produced less PGE2. In summary, these studies demonstrate that T. cruzi infection activates cardiac myocyte iPLA2γ, resulting in increased AA and PGE2 release, mediators that may be essential for host survival during acute infection. Thus, these studies suggest that iPLA2γ plays a cardioprotective role during the acute stage of Chagas' disease.
Collapse
|
46
|
Mukherjee S, Sadekar N, Ashton AW, Huang H, Spray DC, Lisanti MP, Machado FS, Weiss LM, Tanowitz HB. Identification of a functional prostanoid-like receptor in the protozoan parasite, Trypanosoma cruzi. Parasitol Res 2013; 112:1417-25. [PMID: 23403991 DOI: 10.1007/s00436-012-3271-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 12/26/2012] [Indexed: 11/26/2022]
Abstract
Trypanosoma cruzi infection in humans and experimental animals causes Chagas disease which is often accompanied by myocarditis, cardiomyopathy, and vasculopathy. T. cruzi-derived thromboxane A2 (TXA2) modulates vasculopathy and other pathophysiological features of Chagasic cardiomyopathy. Here, we provide evidence that epimastigotes, trypomastigotes, and amastigotes of T. cruzi (Brazil and Tulahuen strains) express a biologically active prostanoid receptor (PR) that is responsive to TXA2 mimetics, e.g. IBOP. This putative receptor, TcPR, is mainly localized in the flagellar membrane of the parasites and shows a similar glycosylation pattern to that of bona fide thromboxane prostanoid (TP) receptors obtained from human platelets. Furthermore, TXA2-PR signal transduction activates T. cruzi-specific MAPK pathways. While mammalian TP is a G-protein coupled receptor (GPCR); T. cruzi genome sequencing has not demonstrated any confirmed GPCRs in these parasites. Based on this genome sequencing it is likely that TcPR is unique in these protists with no counterpart in mammals. TXA2 is a potent vasoconstrictor which contributes to the pathogenesis of Chagasic cardiovascular disease. It may, however, also control parasite differentiation and proliferation in the infected host allowing the infection to progress to a chronic state.
Collapse
Affiliation(s)
- Shankar Mukherjee
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Trypanosoma cruzi infection and endothelin-1 cooperatively activate pathogenic inflammatory pathways in cardiomyocytes. PLoS Negl Trop Dis 2013; 7:e2034. [PMID: 23409199 PMCID: PMC3566987 DOI: 10.1371/journal.pntd.0002034] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 12/12/2012] [Indexed: 12/15/2022] Open
Abstract
Trypanosoma cruzi, the causative agent of Chagas' disease, induces multiple responses in the heart, a critical organ of infection and pathology in the host. Among diverse factors, eicosanoids and the vasoactive peptide endothelin-1 (ET-1) have been implicated in the pathogenesis of chronic chagasic cardiomyopathy. In the present study, we found that T. cruzi infection in mice induces myocardial gene expression of cyclooxygenase-2 (Cox2) and thromboxane synthase (Tbxas1) as well as endothelin-1 (Edn1) and atrial natriuretic peptide (Nppa). T. cruzi infection and ET-1 cooperatively activated the Ca2+/calcineurin (Cn)/nuclear factor of activated T cells (NFAT) signaling pathway in atrial myocytes, leading to COX-2 protein expression and increased eicosanoid (prostaglandins E2 and F2α, thromboxane A2) release. Moreover, T. cruzi infection of ET-1-stimulated cardiomyocytes resulted in significantly enhanced production of atrial natriuretic peptide (ANP), a prognostic marker for impairment in cardiac function of chagasic patients. Our findings support an important role for the Ca2+/Cn/NFAT cascade in T. cruzi-mediated myocardial production of inflammatory mediators and may help define novel therapeutic targets. Chronic cardiomyopathy is the most common and severe manifestation of human Chagas' disease, caused by the protozoan parasite Trypanosoma cruzi. Among diverse inflammation-promoting moieties, eicosanoids and the vasoactive peptide endothelin-1 (ET-1) have been implicated in its pathogenesis. Nevertheless, the link between these two factors has not yet been identified. In the present study, we found that T. cruzi infection induces gene expression of ET-1 and eicosanoid-forming enzymes in the heart of infected mice. We also demonstrated that HL-1 atrial myocytes respond to ET-1 stimulus and T. cruzi infection by induction of cyclooxygenase-2 through activation of the Ca2+/calcineurin/NFAT intracellular signaling pathway. Moreover, the cooperation between T. cruzi and ET-1 leads to overproduction of eicosanoids (prostaglandins E2 and F2α, thromboxane A2) and the pro-hypertrophic atrial natriuretic peptide. Our results support an important role for NFAT in T. cruzi plus ET-1-dependent induction of key agents of pathogenesis in chronic chagasic cardiomyopathy. Identification of the Ca2+/calcineurin/NFAT cascade as mediator of cardiovascular pathology in Chagas' disease advances our understanding of host-parasite interrelationship and may help define novel potential targets for therapeutic interventions to ameliorate or prevent cardiomyopathy during chronic T. cruzi infection.
Collapse
|
48
|
Yang CW, Unanue ER. Neutrophils control the magnitude and spread of the immune response in a thromboxane A2-mediated process. ACTA ACUST UNITED AC 2013; 210:375-87. [PMID: 23337807 PMCID: PMC3570104 DOI: 10.1084/jem.20122183] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Neutrophil-produced thromboxane A2 controls the magnitude and spread of T cell responses to distal lymph nodes. Neutrophils are obligate cells entering lymph nodes shortly after immunization with protein antigens in adjuvants, starting during the first hour and continuing for several days in two distinct waves. Previously, we demonstrated the strong suppressive effects of neutrophils on CD4 T cell and B cell responses, using either neutrophil-depleting antibodies or genetically neutropenic mice. In this study, we find that neutrophils are the major cells controlling the spread of T cell responses to distal lymph nodes. Although in the presence of neutrophils, ∼75% of the response was restricted to the draining node, in their absence, most of the response was found in distal nodes. Prostanoids were responsible for the rapid entry of neutrophils into the draining nodes, as well as for the two distinct neutrophil effects: the modulation of the magnitude of the cellular response, and in its spread outside the draining nodes. Neutrophil-produced thromboxane A2 was the key eicosanoid controlling both effects. Adoptive transfer of neutrophils into mice genetically deficient in neutrophils indicated their role in both. These functions of neutrophils are important in infections and vaccinations with adjuvants where neutrophils are abundant in the initial stages.
Collapse
Affiliation(s)
- Chiao-Wen Yang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
49
|
Rodríguez-Angulo H, Toro-Mendoza J, Marques J, Bonfante-Cabarcas R, Mijares A. Induction of chagasic-like arrhythmias in the isolated beating hearts of healthy rats perfused with Trypanosoma cruzi-conditioned medium. ACTA ACUST UNITED AC 2013; 46:58-64. [PMID: 23314340 PMCID: PMC3854352 DOI: 10.1590/1414-431x20122409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 09/03/2012] [Indexed: 11/21/2022]
Abstract
Chagas' myocardiopathy, caused by the intracellular protozoan Trypanosoma cruzi, is characterized by microvascular alterations, heart failure and arrhythmias. Ischemia and arrythmogenesis have been attributed to proteins shed by the parasite, although this has not been fully demonstrated. The aim of the present investigation was to study the effect of substances shed by T. cruzi on ischemia/reperfusion-induced arrhythmias. We performed a triple ischemia-reperfusion (I/R) protocol whereby the isolated beating rat hearts were perfused with either Vero-control or Vero T. cruzi-infected conditioned medium during the different stages of ischemia and subsequently reperfused with Tyrode's solution. ECG and heart rate were recorded during the entire experiment. We observed that triple I/R-induced bradycardia was associated with the generation of auricular-ventricular blockade during ischemia and non-sustained nodal and ventricular tachycardia during reperfusion. Interestingly, perfusion with Vero-infected medium produced a delay in the reperfusion-induced recovery of heart rate, increased the frequency of tachycardic events and induced ventricular fibrillation. These results suggest that the presence of parasite-shed substances in conditioned media enhances the arrhythmogenic effects that occur during the I/R protocol.
Collapse
Affiliation(s)
- H Rodríguez-Angulo
- Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | | | | | | | | |
Collapse
|
50
|
Machado FS, Dutra WO, Esper L, Gollob K, Teixeira MM, Factor SM, Weiss LM, Nagajyothi F, Tanowitz HB, Garg NJ. Current understanding of immunity to Trypanosoma cruzi infection and pathogenesis of Chagas disease. Semin Immunopathol 2012; 34:753-70. [PMID: 23076807 PMCID: PMC3498515 DOI: 10.1007/s00281-012-0351-7] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 09/21/2012] [Indexed: 02/06/2023]
Abstract
Chagas disease caused by Trypanosoma cruzi remains an important neglected tropical disease and a cause of significant morbidity and mortality. No longer confined to endemic areas of Latin America, it is now found in non-endemic areas due to immigration. The parasite may persist in any tissue, but in recent years, there has been increased recognition of adipose tissue both as an early target of infection and a reservoir of chronic infection. The major complications of this disease are cardiomyopathy and megasyndromes involving the gastrointestinal tract. The pathogenesis of Chagas disease is complex and multifactorial involving many interactive pathways. The significance of innate immunity, including the contributions of cytokines, chemokines, reactive oxygen species, and oxidative stress, has been emphasized. The role of the components of the eicosanoid pathway such as thromboxane A(2) and the lipoxins has been demonstrated to have profound effects as both pro- and anti-inflammatory factors. Additionally, we discuss the vasoconstrictive actions of thromboxane A(2) and endothelin-1 in Chagas disease. Human immunity to T. cruzi infection and its role in pathogen control and disease progression have not been fully investigated. However, recently, it was demonstrated that a reduction in the anti-inflammatory cytokine IL-10 was associated with clinically significant chronic chagasic cardiomyopathy.
Collapse
Affiliation(s)
- Fabiana S. Machado
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walderez O. Dutra
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Departments of Microbiology and Immunology and Pathology, Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX
| | - Lisia Esper
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Kenneth Gollob
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Santa Casa Hospital, Belo Horizonte, Brazil
- SRI International, Biosciences Division, Menlo Park, CA
- National Institute of Science and Technology in Tropical Diseases, Belo Horizonte, MG, Brazil
| | - Mauro M. Teixeira
- Departments of Biochemistry and Immunology and Morphology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Stephen M. Factor
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Louis M. Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Fnu Nagajyothi
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
| | - Herbert B. Tanowitz
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Nisha J. Garg
- Departments of Microbiology and Immunology and Pathology, Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX
| |
Collapse
|