1
|
Khan AA, Taylor MC, Fortes Francisco A, Jayawardhana S, Atherton RL, Olmo F, Lewis MD, Kelly JM. Animal models for exploring Chagas disease pathogenesis and supporting drug discovery. Clin Microbiol Rev 2024; 37:e0015523. [PMID: 39545730 PMCID: PMC11629624 DOI: 10.1128/cmr.00155-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024] Open
Abstract
SUMMARYInfections with the parasitic protozoan Trypanosoma cruzi cause Chagas disease, which results in serious cardiac and/or digestive pathology in 30%-40% of individuals. However, symptomatic disease can take decades to become apparent, and there is a broad spectrum of possible outcomes. The complex and long-term nature of this infection places a major constraint on the scope for experimental studies in humans. Accordingly, predictive animal models have been a mainstay of Chagas disease research. The resulting data have made major contributions to our understanding of parasite biology, immune responses, and disease pathogenesis and have provided a platform that informs and facilitates the global drug discovery effort. Here, we provide an overview of available animal models and illustrate how they have had a key impact across the field.
Collapse
Affiliation(s)
- Archie A. Khan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Martin C. Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Amanda Fortes Francisco
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shiromani Jayawardhana
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Richard L. Atherton
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Francisco Olmo
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Michael D. Lewis
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - John M. Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
2
|
do Carmo Neto JR, Braga YLL, Franco PIR, de Oliveira JF, Trevisan RO, Mendes KM, de Oliveira MAP, Celes MRN, Silva ACA, Machado JR, da Silva MV. Achieving the Optimal AgO Concentrations to Modulate the Anti- Trypanosoma cruzi Activity of Ag-ZnO/AgO Nanocomposites: In Vivo Investigations. Pharmaceutics 2024; 16:1415. [PMID: 39598539 PMCID: PMC11597568 DOI: 10.3390/pharmaceutics16111415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: For the development of new treatments, the acute phase of Chagas disease (CD) in experimental models acts as a filter to screen out potentially effective interventions. Therefore, the aim of this study was to evaluate ZnO nanocrystals and Ag-ZnO/AgO nanocomposites containing different proportions of silver (ZnO:5Ag, ZnO:9Ag and ZnO:11Ag) in an experimental model of the acute phase of CD. Methods: C57Bl/6 mice were infected with 1000 forms of the Colombian strain of T. cruzi. The treatment was carried out by gavage with 5 mg/kg/d for 7 consecutive days from the first detection of parasitemia. Weight, parasitemia and survival were assessed during treatment and up to the day of euthanasia. After euthanasia, the cardiac and intestinal parasitism, inflammatory infiltrate, collagen deposition and cytokine dosages were analyzed. Results: It was observed that the nanocomposites ZnO:9Ag and ZnO:11Ag were the most effective in reducing parasitemia and increasing the survival of the infected animals. However, pure ZnO induced the maintenance of parasitemia and reduced their survival. The ZnO:9Ag and ZnO:11Ag nanocomposites were able to reduce the number of cardiac amastigote nests. In addition, they were responsible for reducing TNF-α and IL-6 in situ. ZnO:9Ag and ZnO:11Ag induced a reduction in the intestinal inflammatory infiltrate and neuronal protection in the myenteric plexus, as well as reducing TNF-α in situ. Conclusions: Based on these results, it is suggested that there is an ideal concentration in terms of the proportion of Ag/AgO and ZnO in nanocomposites for use against CD. Thus, ZnO:9Ag or ZnO:11Ag nanomaterials are potential candidates for the development of new biotechnological products for the therapy of CD.
Collapse
Affiliation(s)
- José Rodrigues do Carmo Neto
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia 74605-050, GO, Brazil; (J.R.d.C.N.); (Y.L.L.B.); (P.I.R.F.); (J.F.d.O.); (M.A.P.d.O.); (M.R.N.C.)
| | - Yarlla Loyane Lira Braga
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia 74605-050, GO, Brazil; (J.R.d.C.N.); (Y.L.L.B.); (P.I.R.F.); (J.F.d.O.); (M.A.P.d.O.); (M.R.N.C.)
| | - Pablo Igor Ribeiro Franco
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia 74605-050, GO, Brazil; (J.R.d.C.N.); (Y.L.L.B.); (P.I.R.F.); (J.F.d.O.); (M.A.P.d.O.); (M.R.N.C.)
| | - Jordana Fernandes de Oliveira
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia 74605-050, GO, Brazil; (J.R.d.C.N.); (Y.L.L.B.); (P.I.R.F.); (J.F.d.O.); (M.A.P.d.O.); (M.R.N.C.)
| | - Rafael Obata Trevisan
- Department of General Pathology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil; (R.O.T.); (K.M.M.)
| | - Karen Martins Mendes
- Department of General Pathology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil; (R.O.T.); (K.M.M.)
| | - Milton Adriano Pelli de Oliveira
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia 74605-050, GO, Brazil; (J.R.d.C.N.); (Y.L.L.B.); (P.I.R.F.); (J.F.d.O.); (M.A.P.d.O.); (M.R.N.C.)
| | - Mara Rúbia Nunes Celes
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia 74605-050, GO, Brazil; (J.R.d.C.N.); (Y.L.L.B.); (P.I.R.F.); (J.F.d.O.); (M.A.P.d.O.); (M.R.N.C.)
| | - Anielle Christine Almeida Silva
- Laboratório de Novos Materiais Nanoestruturados e Funcionais (LNMIS), Physics Institute, Federal University of Alagoas, Maceió 57072-900, AL, Brazil;
| | - Juliana Reis Machado
- Department of Bioscience and Technology, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia 74605-050, GO, Brazil; (J.R.d.C.N.); (Y.L.L.B.); (P.I.R.F.); (J.F.d.O.); (M.A.P.d.O.); (M.R.N.C.)
- Department of General Pathology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil; (R.O.T.); (K.M.M.)
| | - Marcos Vinícius da Silva
- Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences of Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil;
| |
Collapse
|
3
|
de Alba-Alvarado MC, Cabrera-Bravo M, Zenteno E, Salazar-Schetino PM, Bucio-Torres MI. The Functions of Cytokines in the Cardiac Immunopathogenesis of Chagas Disease. Pathogens 2024; 13:870. [PMID: 39452741 PMCID: PMC11510034 DOI: 10.3390/pathogens13100870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024] Open
Abstract
Chagas disease is a complex zoonosis. Clinically, it presents in two distinct phases, acute and chronic. The ability of patients to respond to Trypanosoma cruzi infection depends on the balance between inflammatory and anti-inflammatory responses, in which cytokines play a key regulatory role. In this review, we discuss the role of cytokines in regulating the host response and as mediators of cardiac injury by inducing profibrotic alterations. The importance of characterizing cytokine profiles as biomarkers of the evolution of cardiac damage in T.-cruzi-infected individuals is also emphasized.
Collapse
Affiliation(s)
- Mariana Citlalli de Alba-Alvarado
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad México 04510, Mexico; (M.C.-B.); (P.M.S.-S.)
| | - Margarita Cabrera-Bravo
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad México 04510, Mexico; (M.C.-B.); (P.M.S.-S.)
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico;
| | - Paz María Salazar-Schetino
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad México 04510, Mexico; (M.C.-B.); (P.M.S.-S.)
| | - Martha Irene Bucio-Torres
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad México 04510, Mexico; (M.C.-B.); (P.M.S.-S.)
| |
Collapse
|
4
|
Cristovão-Silva AC, Brelaz-de-Castro MCA, Dionisio da Silva E, Leite ACL, Santiago LBAA, Conceição JMD, da Silva Tiburcio R, de Santana DP, Bedor DCG, de Carvalho BÍV, Ferreira LFGR, de Freitas E Silva R, Alves Pereira VR, Hernandes MZ. Trypanosoma cruzi killing and immune response boosting by novel phenoxyhydrazine-thiazole against Chagas disease. Exp Parasitol 2024; 261:108749. [PMID: 38593864 DOI: 10.1016/j.exppara.2024.108749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/23/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
Trypanosoma cruzi (T. cruzi) causes Chagas, which is a neglected tropical disease (NTD). WHO estimates that 6 to 7 million people are infected worldwide. Current treatment is done with benznidazole (BZN), which is very toxic and effective only in the acute phase of the disease. In this work, we designed, synthesized, and characterized thirteen new phenoxyhydrazine-thiazole compounds and applied molecular docking and in vitro methods to investigate cell cytotoxicity, trypanocide activity, nitric oxide (NO) production, cell death, and immunomodulation. We observed a higher predicted affinity of the compounds for the squalene synthase and 14-alpha demethylase enzymes of T. cruzi. Moreover, the compounds displayed a higher predicted affinity for human TLR2 and TLR4, were mildly toxic in vitro for most mammalian cell types tested, and LIZ531 (IC50 2.8 μM) was highly toxic for epimastigotes, LIZ311 (IC50 8.6 μM) for trypomastigotes, and LIZ331 (IC50 1.9 μM) for amastigotes. We observed that LIZ311 (IC50 2.5 μM), LIZ431 (IC50 4.1 μM) and LIZ531 (IC50 5 μM) induced 200 μg/mL of NO and JM14 induced NO production in three different concentrations tested. The compound LIZ331 induced the production of TNF and IL-6. LIZ311 induced the secretion of TNF, IFNγ, IL-2, IL-4, IL-10, and IL-17, cell death by apoptosis, decreased acidic compartment formation, and induced changes in the mitochondrial membrane potential. Taken together, LIZ311 is a promising anti-T. cruzi compound is not toxic to mammalian cells and has increased antiparasitic activity and immunomodulatory properties.
Collapse
Affiliation(s)
- Ana Catarina Cristovão-Silva
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil
| | - Maria Carolina Accioly Brelaz-de-Castro
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil; Laboratory of Parasitology, Vitória Academic Center, Federal University of Pernambuco, 55608-680, Vitória de Santo Antão, Pernambuco, Brazil
| | - Elis Dionisio da Silva
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil
| | - Ana Cristina Lima Leite
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Lizandra Beatriz Amorim Alves Santiago
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Juliana Maria da Conceição
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Robert da Silva Tiburcio
- Laboratory of Planning and Synthesis in Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Davi Pereira de Santana
- Pharmaceutical and Cosmetic Development Center (NUDFAC), Department of Pharmaceutical Science, Federal University of Pernambuco, Recife, PE, Brazil
| | - Danilo Cesar Galindo Bedor
- Pharmaceutical and Cosmetic Development Center (NUDFAC), Department of Pharmaceutical Science, Federal University of Pernambuco, Recife, PE, Brazil
| | - Breno Ítalo Valença de Carvalho
- Pharmaceutical and Cosmetic Development Center (NUDFAC), Department of Pharmaceutical Science, Federal University of Pernambuco, Recife, PE, Brazil
| | - Luiz Felipe Gomes Rebello Ferreira
- Laboratory of Theoretical and Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil
| | - Rafael de Freitas E Silva
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil.
| | - Valéria Rêgo Alves Pereira
- Laboratory of Immunopathology and Molecular Biology, Aggeu Magalhães Institute, Oswaldo Cruz Foundation, 50670-42, Recife, Pernambuco, Brazil.
| | - Marcelo Zaldini Hernandes
- Laboratory of Theoretical and Medicinal Chemistry, Pharmaceutical Sciences Department, Federal University of Pernambuco, 50740-520, Recife, Pernambuco, Brazil.
| |
Collapse
|
5
|
Tran DT, Batchu SN, Advani A. Interferons and interferon-related pathways in heart disease. Front Cardiovasc Med 2024; 11:1357343. [PMID: 38665231 PMCID: PMC11043610 DOI: 10.3389/fcvm.2024.1357343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Interferons (IFNs) and IFN-related pathways play key roles in the defence against microbial infection. However, these processes may also be activated during the pathogenesis of non-infectious diseases, where they may contribute to organ injury, or function in a compensatory manner. In this review, we explore the roles of IFNs and IFN-related pathways in heart disease. We consider the cardiac effects of type I IFNs and IFN-stimulated genes (ISGs); the emerging role of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway; the seemingly paradoxical effects of the type II IFN, IFN-γ; and the varied actions of the interferon regulatory factor (IRF) family of transcription factors. Recombinant IFNs and small molecule inhibitors of mediators of IFN receptor signaling are already employed in the clinic for the treatment of some autoimmune diseases, infections, and cancers. There has also been renewed interest in IFNs and IFN-related pathways because of their involvement in SARS-CoV-2 infection, and because of the relatively recent emergence of cGAS-STING as a pattern recognition receptor-activated pathway. Whether these advances will ultimately result in improvements in the care of those experiencing heart disease remains to be determined.
Collapse
Affiliation(s)
| | | | - Andrew Advani
- Keenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
6
|
Liu K, Han B. Role of immune cells in the pathogenesis of myocarditis. J Leukoc Biol 2024; 115:253-275. [PMID: 37949833 DOI: 10.1093/jleuko/qiad143] [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: 08/15/2023] [Revised: 10/15/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
Myocarditis is an inflammatory heart disease that mostly affects young people. Myocarditis involves a complex immune network; however, its detailed pathogenesis is currently unclear. The diversity and plasticity of immune cells, either in the peripheral blood or in the heart, have been partially revealed in a number of previous studies involving patients and several kinds of animal models with myocarditis. It is the complexity of immune cells, rather than one cell type that is the culprit. Thus, recognizing the individual intricacies within immune cells in the context of myocarditis pathogenesis and finding the key intersection of the immune network may help in the diagnosis and treatment of this condition. With the vast amount of cell data gained on myocarditis and the recent application of single-cell sequencing, we summarize the multiple functions of currently recognized key immune cells in the pathogenesis of myocarditis to provide an immune background for subsequent investigations.
Collapse
Affiliation(s)
- Keyu Liu
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital, Shandong University, Cheeloo Colledge of Medicine, No. 324 Jingwu Road, 250021, Jinan, China
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, 250021, Jinan, China
- Shandong Provincial Hospital, Shandong Provincial Clinical Research Center for Children' s Health and Disease office, No. 324 Jingwu Road, 250021, Jinan, China
| |
Collapse
|
7
|
Vellozo NS, Matos-Silva TC, Lopes MF. Immunopathogenesis in Trypanosoma cruzi infection: a role for suppressed macrophages and apoptotic cells. Front Immunol 2023; 14:1244071. [PMID: 37662946 PMCID: PMC10469960 DOI: 10.3389/fimmu.2023.1244071] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
During Trypanosoma cruzi infection, macrophages phagocytose parasites and remove apoptotic cells through efferocytosis. While macrophage 1 (M1) produces proinflammatory cytokines and NO and fights infection, M2 macrophages are permissive host cells that express arginase 1 and play a role in tissue repair. The regulation of M1 and M2 phenotypes might either induce or impair macrophage-mediated immunity towards parasite control or persistence in chronic Chagas disease. Here, we highlight a key role of macrophage activation in early immune responses to T. cruzi that prevent escalating parasitemia, heart parasitism, and mortality during acute infection. We will discuss the mechanisms of macrophage activation and deactivation, such as T cell cytokines and efferocytosis, and how to improve macrophage-mediated immunity to prevent parasite persistence, inflammation, and the development of chagasic cardiomyopathy. Potential vaccines or therapy must enhance early T cell-macrophage crosstalk and parasite control to restrain the pathogenic outcomes of parasite-induced inflammation in the heart.
Collapse
Affiliation(s)
| | | | - Marcela F. Lopes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
8
|
Gómez-Ochoa SA, Bautista-Niño PK, Rojas LZ, Hunziker L, Muka T, Echeverría LE. Circulating MicroRNAs and myocardial involvement severity in chronic Chagas cardiomyopathy. Front Cell Infect Microbiol 2022; 12:922189. [PMID: 36004323 PMCID: PMC9393411 DOI: 10.3389/fcimb.2022.922189] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/18/2022] [Indexed: 12/18/2022] Open
Abstract
Background Chronic Chagas Cardiomyopathy (CCM) is characterized by a unique pathophysiology in which inflammatory, microvascular and neuroendocrine processes coalesce in the development of one of the most severe cardiomyopathies affecting humans. Despite significant advances in understanding the molecular mechanisms involved in this disease, scarce information is available regarding microRNAs and clinical parameters of disease severity. We aimed to evaluate the association between circulating levels of six microRNAs with markers of myocardial injury and prognosis in this population. Methods Patients with CCM and reduced ejection fraction were included in a prospective exploratory cohort study. We assessed the association of natural log-transformed values of six circulating microRNAs (miR-34a-5p, miR-208a-5p, miR-185-5p, miR-223-5p, let-7d-5p, and miR-454-5p) with NT-proBNP levels and echocardiographic variables using linear regression models adjusted for potential confounders. By using Cox Proportional Hazard models, we examined whether levels of microRNAs could predict a composite outcome (CO), including all-cause mortality, cardiac transplantation, and implantation of a left ventricular assist device (LVAD). Finally, for mRNAs showing significant associations, we predicted the target genes and performed pathway analyses using Targetscan and Reactome Pathway Browser. Results Seventy-four patients were included (59% males, median age: 64 years). After adjustment for age, sex, body mass index, and heart failure medications, only increasing miR-223-5p relative expression levels were significantly associated with better myocardial function markers, including left atrium area (Coef. -10.2; 95% CI -16.35; -4.09), end-systolic (Coef. -45.3; 95% CI -74.06; -16.61) and end-diastolic volumes (Coef. -46.1; 95% CI -81.99; -10.26) of the left ventricle. Moreover, we observed that higher miR-223-5p levels were associated with better left-ventricle ejection fraction and lower NT-proBNP levels. No associations were observed between the six microRNAs and the composite outcome. A total of 123 target genes for miR-223-5p were obtained. From these, several target pathways mainly related to signaling by receptor tyrosine kinases were identified. Conclusions The present study found an association between miR-223-5p and clinical parameters of CCM, with signaling pathways related to receptor tyrosine kinases as a potential mechanism linking low levels of miR-223-5p with CCM worsening.
Collapse
Affiliation(s)
| | | | - Lyda Z. Rojas
- Research Group and Development of Nursing Knowledge (GIDCEN-FCV), Research Center, Fundación Cardiovascular de Colombia, Floridablanca, Colombia
| | - Lukas Hunziker
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Taulant Muka
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- *Correspondence: Taulant Muka,
| | - Luis E. Echeverría
- Heart Failure and Heart Transplant Clinic, Fundación Cardiovascular de Colombia, Floridablanca, Colombia
| |
Collapse
|
9
|
Vieira RDS, Nascimento MS, Noronha IH, Vasconcelos JRC, Benvenuti LA, Barber GN, Câmara NOS, Kalil J, Cunha-Neto E, Almeida RR. STING Signaling Drives Production of Innate Cytokines, Generation of CD8 + T Cells and Enhanced Protection Against Trypanosoma cruzi Infection. Front Immunol 2022; 12:775346. [PMID: 35095849 PMCID: PMC8795786 DOI: 10.3389/fimmu.2021.775346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022] Open
Abstract
A variety of signaling pathways are involved in the induction of innate cytokines and CD8+ T cells, which are major players in protection against acute Trypanosoma cruzi infection. Previous data have demonstrated that a TBK-1/IRF3-dependent signaling pathway promotes IFN-β production in response to Trypanosoma cruzi, but the role for STING, a main interactor of these proteins, remained to be addressed. Here, we demonstrated that STING signaling is required for production of IFN-β, IL-6, and IL-12 in response to Trypanosoma cruzi infection and that STING absence negatively impacts activation of IRF-dependent pathways in response to the parasite. We reported no significant activation of IRF-dependent pathways and cytokine expression in RAW264.7 macrophages in response to heat-killed trypomastigotes. In addition, we showed that STING is essential for T. cruzi DNA-mediated induction of IFN-β, IL-6, and IL-12 gene expression in RAW264.7 macrophages. We demonstrated that STING-knockout mice have significantly higher parasitemia from days 5 to 8 of infection and higher heart parasitism at day 13 after infection. Although we observed similar heart inflammatory infiltrates at day 13 after infection, IFN-β, IL-12, CXCL9, IFN-γ, and perforin gene expression were lower in the absence of STING. We also showed an inverse correlation between parasite DNA and the expression of CXCL9, IFN-γ, and perforin genes in the hearts of infected animals at day 13 after infection. Finally, we reported that STING signaling is required for splenic IFN-β and IL-6 expression early after infection and that STING deficiency results in lower numbers of splenic parasite-specific IFN-γ and IFN-γ/perforin-producing CD8+ T cells, indicating a pivotal role for STING signaling in immunity to Trypanosoma cruzi.
Collapse
Affiliation(s)
- Raquel de Souza Vieira
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marilda Savoia Nascimento
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Isaú Henrique Noronha
- Laboratório de Vacinas Recombinantes, Departamento de Biociências, Universidade Federal de São Paulo, Santos, Brazil
| | | | - Luiz Alberto Benvenuti
- Divisão de Patologia, Instituto do Coração (INCOR), Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Glen N Barber
- Department of Cell Biology, University of Miami, Miami, FL, United States
| | - Niels Olsen Saraiva Câmara
- Laboratório de Imunologia Experimental e Clínica, Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Laboratório de Imunologia de Transplantes, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Jorge Kalil
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto de Investigação em Imunologia (III), Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.,Instituto de Investigação em Imunologia (III), Instituto Nacional de Ciência e Tecnologia (INCT), São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratório de Imunologia, Instituto do Coração, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Braga YLL, Neto JRC, Costa AWF, Silva MVT, Silva MV, Celes MRN, Oliveira MAP, Joosten LAB, Ribeiro-Dias F, Gomes RS, Machado JR. Interleukin-32 γ in the Control of Acute Experimental Chagas Disease. J Immunol Res 2022; 2022:7070301. [PMID: 35097133 PMCID: PMC8794684 DOI: 10.1155/2022/7070301] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
Chagas disease (CD) is an important parasitic disease caused by Trypanosoma cruzi. Interleukin-32 (IL-32) plays an important role in inflammation and in the development of Th1/Th17 acquired immune responses. We evaluated the influence of IL-32γ on the immune response profile, pathogenesis of myocarditis in acute experimental CD, and control of the disease. For this, C57BL/6 wild-type (WT) and IL-32γTg mice were infected subcutaneously with 1,000 forms of Colombian strain of T. cruzi. In the histopathological analyzes, T. cruzi nests, myocarditis, and collagen were quantified in cardiac tissue. Cytokine productions (IL-32, IFN-γ, TNF-α, IL-10, and IL-17) were measured in cardiac homogenate by ELISA. The IL-32γTg mice showed a better control of parasitemia and T. cruzi nests in the heart than WT mice. Infected-WT and -IL-32γTg mice showed similar levels of IFN-γ, TNF-α, and IL-17, but IL-10 was significantly higher expressed in IL-32γTg than in WT mice. The cytokine profile found in IL-32γTg animals contributed to body weight maintenance, parasitemia control, and survival. Our results indicate that the presence of human IL-32γ in mice infected with the Colombian strain of T. cruzi is important for infection control during the acute phase of Chagas disease.
Collapse
Affiliation(s)
- Yarlla L. L. Braga
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - José R. C. Neto
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Arthur W. F. Costa
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Muriel V. T. Silva
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Marcos V. Silva
- Departamento de Microbiologia, Bioquímica e Imunologia, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Mara R. N. Celes
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Milton A. P. Oliveira
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Leo A. B. Joosten
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
- Department of Internal Medicine and Radboud Center of Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Fátima Ribeiro-Dias
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Rodrigo S. Gomes
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Juliana R. Machado
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
- Departamento de Patologia, Genética e Evolução, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| |
Collapse
|
11
|
Santos-Miranda A, Costa AD, Joviano-Santos JV, Rhana P, Bruno AS, Rocha P, Cau SB, Vieira LQ, Cruz JS, Roman-Campos D. Inhibition of calcium/calmodulin (Ca 2+ /CaM)-Calcium/calmodulin-dependent protein kinase II (CaMKII) axis reduces in vitro and ex vivo arrhythmias in experimental Chagas disease. FASEB J 2021; 35:e21901. [PMID: 34569665 DOI: 10.1096/fj.202101060r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/11/2022]
Abstract
Chagasic cardiomyopathy (CCC) is one of the main causes of heart failure and sudden death in Latin America. To date, there is no available medication to prevent or reverse the onset of cardiac symptoms. CCC occurs in a scenario of disrupted calcium dynamics and enhanced oxidative stress, which combined, may favor the hyper activation of calcium/calmodulin (Ca2+ /CaM)-calcium/calmodulin-dependent protein kinase II (CaMKII) (Ca2+ /CaM-CaMKII) pathway, which is fundamental for heart physiology and it is implicated in other cardiac diseases. Here, we evaluated the association between Ca2+ /CaM-CaMKII in the electro-mechanical (dys)function of the heart in the early stage of chronic experimental Trypanosoma cruzi infection. We observed that in vitro and ex vivo inhibition of Ca2+ /CaM-CaMKII reversed the arrhythmic profile of isolated hearts and isolated left-ventricles cardiomyocytes. The benefits of the limited Ca2+ /CaM-CaMKII activation to cardiomyocytes' electrical properties are partially related to the restoration of Ca2+ dynamics in a damaged cellular environment created after T. cruzi infection. Moreover, Ca2+ /CaM-CaMKII inhibition prevented the onset of arrhythmic contractions on isolated heart preparations of chagasic mice and restored the responsiveness to the increase in the left-ventricle pre-load. Taken together, our data provide the first experimental evidence for the potential of targeting Ca2+ /CaM-CaMKII pathway as a novel therapeutic target to treat CCC.
Collapse
Affiliation(s)
| | - Alexandre D Costa
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Paula Rhana
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alexandre Santos Bruno
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Peter Rocha
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Stefany Bruno Cau
- Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leda Q Vieira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jader S Cruz
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danilo Roman-Campos
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Zuma AA, Dos Santos Barrias E, de Souza W. Basic Biology of Trypanosoma cruzi. Curr Pharm Des 2021; 27:1671-1732. [PMID: 33272165 DOI: 10.2174/1381612826999201203213527] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022]
Abstract
The present review addresses basic aspects of the biology of the pathogenic protozoa Trypanosoma cruzi and some comparative information of Trypanosoma brucei. Like eukaryotic cells, their cellular organization is similar to that of mammalian hosts. However, these parasites present structural particularities. That is why the following topics are emphasized in this paper: developmental stages of the life cycle in the vertebrate and invertebrate hosts; the cytoskeleton of the protozoa, especially the sub-pellicular microtubules; the flagellum and its attachment to the protozoan body through specialized junctions; the kinetoplast-mitochondrion complex, including its structural organization and DNA replication; glycosome and its role in the metabolism of the cell; acidocalcisome, describing its morphology, biochemistry, and functional role; cytostome and the endocytic pathway; the organization of the endoplasmic reticulum and Golgi complex; the nucleus, describing its structural organization during interphase and division; and the process of interaction of the parasite with host cells. The unique characteristics of these structures also make them interesting chemotherapeutic targets. Therefore, further understanding of cell biology aspects contributes to the development of drugs for chemotherapy.
Collapse
Affiliation(s)
- Aline A Zuma
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Emile Dos Santos Barrias
- Laboratorio de Metrologia Aplicada a Ciencias da Vida, Diretoria de Metrologia Aplicada a Ciencias da Vida - Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Rio de Janeiro, Brazil
| | - Wanderley de Souza
- Laboratorio de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho - Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Jimenes DR, Teixeira Junior NR, Pereira AV, Berti JA, Barbosa CP, Sant'Ana DDMG. Human apoCIII transgenic mice with epicardial adipose tissue inflammation and PRESERVATION of the cardiac plexus. Exp Gerontol 2021; 148:111261. [PMID: 33647361 DOI: 10.1016/j.exger.2021.111261] [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: 02/26/2020] [Revised: 12/16/2020] [Accepted: 01/24/2021] [Indexed: 10/22/2022]
Abstract
Hypertriglyceridemia is a result of the increase in the serum levels of lipoproteins, which are responsible for the transport of triglycerides and can be caused by genetic and/or metabolic factors. Animal models which either express or lack genes related to changes in the lipoproteins profile are useful to understand lipid metabolism. Apolipoprotein CIII (apoCIII) is an important modulator of hepatic production and peripheral removal of triglycerides. Mice that overexpress the apoCIII gene become hypertriglyceridemic, showing high concentrations of free fatty acids in the blood. Since hypertriglyceridemia is related to atherosclerosis, and the latter refers to cardiac alterations, this study aimed at evaluating the morphological, morphometric and quantitative profiles of the cardiac plexus, as well as the morphometric and histopathological aspects of the epicardial adipose tissue in human apoCIII transgenic mice. Therefore, 8-12-month-old male C57BL/6 mice that overexpressed human apoCIII (CIII) and their respective controls were used. Our results showed that overexpression of human apoCIII did not modify morphological or quantitative parameters of cardiac plexus neurons; however, age increased both, the area and the number of such cells. Furthermore, there was a direct correlation of this dyslipidemia to the thickening of periganglionar type 1 collagens. On the other hand, this overexpression caused epicardial adipose tissue inflammation and an increase in the area of the adipocytes, thus, favoring the recruitment of inflammatory cells in this tissue. In conclusion, this overexpression is harmful since it is related to an increase in cardiac adiposity, as well as to a predisposition to an inflammatory environment in the epicardial fat and to the incidence of cardiovascular diseases.
Collapse
Affiliation(s)
- Diogo Rodrigues Jimenes
- Program of Graduate Studies in Bioscience and Physiopathology - State University of Maringá (PBF-UEM), Brazil.
| | | | | | | | | | - Débora de Mello Gonçales Sant'Ana
- Program of Graduate Studies in Bioscience and Physiopathology - State University of Maringá (PBF-UEM), Brazil; Department of Physiological Sciences (DFS-UEM), Brazil
| |
Collapse
|
14
|
Pérez‐Mazliah D, Ward AI, Lewis MD. Host-parasite dynamics in Chagas disease from systemic to hyper-local scales. Parasite Immunol 2021; 43:e12786. [PMID: 32799361 PMCID: PMC11475410 DOI: 10.1111/pim.12786] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022]
Abstract
Trypanosoma cruzi is a remarkably versatile parasite. It can parasitize almost any nucleated cell type and naturally infects hundreds of mammal species across much of the Americas. In humans, it is the cause of Chagas disease, a set of mainly chronic conditions predominantly affecting the heart and gastrointestinal tract, which can progress to become life threatening. Yet around two thirds of infected people are long-term asymptomatic carriers. Clinical outcomes depend on many factors, but the central determinant is the nature of the host-parasite interactions that play out over the years of chronic infection in diverse tissue environments. In this review, we aim to integrate recent developments in the understanding of the spatial and temporal dynamics of T. cruzi infections with established and emerging concepts in host immune responses in the corresponding phases and tissues.
Collapse
Affiliation(s)
- Damián Pérez‐Mazliah
- York Biomedical Research InstituteHull York Medical SchoolUniversity of YorkYorkUK
| | - Alexander I. Ward
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
| | - Michael D. Lewis
- Department of Infection BiologyFaculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
15
|
Participation of Central Muscarinic Receptors on the Nervous Form of Chagas Disease in Mice Infected via Intracerebroventricular with Colombian Trypanosoma cruzi Strain. Pathogens 2021; 10:pathogens10020121. [PMID: 33503848 PMCID: PMC7922850 DOI: 10.3390/pathogens10020121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 11/30/2022] Open
Abstract
Acute chagasic encephalitis is a clinically severe central nervous system (CNS) manifestation. However, the knowledge of the nervous form of Chagas disease is incomplete. The role of the muscarinic acetylcholine receptor (mAChR) on mice behavior and brain lesions induced by Trypanosoma cruzi (Colombian strain) was herein investigated in mice treated with the mAChR agonist and antagonist (carbachol and atropine), respectively. Immunosuppressed or non-immunosuppressed mice were intracerebroventricularly (icv) or intraperitoneally (ip) infected. All groups were evaluated 15 d.p.i. (days post infection). Intraperitoneally infected animals had subpatent parasitemia. Patent parasitemia occurred only in icv infected mice. The blockade of mAChR increased the parasitemia, parasitism and lesions compared to its activation. Infected not treated (INT ip) mice did not present meningitis and encephalitis, regardless of immunosuppression. INT icv brains presented higher cellularity, discrete signs of cellular degeneration, frequent presence of parasites and focal meningitis. The immunosuppressed atropine + icv mice presented increased intracellular parasitism associated with degenerative parenchymal changes, while carbachol + icv mice presented discrete meningitis, preservation of the cortex and absence of relevant parasitism. Cholinergic receptor blockage increased impairment of coordination vs. receptor activation. Muscarinic cholinergic pathway seems to be involved in immune mediated cell invasion events while its blockade favored infection evolution, brain lesions, and behavioral alterations.
Collapse
|
16
|
Frade-Barros AF, Ianni BM, Cabantous S, Pissetti CW, Saba B, Lin-Wang HT, Buck P, Marin-Neto JA, Schmidt A, Dias F, Hirata MH, Sampaio M, Fragata A, Pereira AC, Donadi E, Rodrigues V, Kalil J, Chevillard C, Cunha-Neto E. Polymorphisms in Genes Affecting Interferon-γ Production and Th1 T Cell Differentiation Are Associated With Progression to Chagas Disease Cardiomyopathy. Front Immunol 2020; 11:1386. [PMID: 32733459 PMCID: PMC7358543 DOI: 10.3389/fimmu.2020.01386] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Chagas disease, caused by the protozoan Trypanosoma cruzi, is endemic in Latin America. Thirty percent of infected individuals develop chronic Chagas cardiomyopathy (CCC), an inflammatory dilated cardiomyopathy that is the most important clinical consequence of T. cruzi infection, while the others remain asymptomatic (ASY). IFN-γ and IFN-γ-producing Th1-type T cells are increased in peripheral blood and CCC myocardium as compared to ASY patients, while the Th1-antagonizing cytokine IL-10 is more expressed in ASY patients. Importantly IFN-γ-producing Th1-type T cells are the most frequent cytokine-producing T cell subset in CCC myocardium, while expression of Th1-antagonizing cytokines IL-10 and IL-4 is unaltered. The control of IFN-γ production by Th1-type T cells may be a key event for progression toward CCC. A genetic component to disease progression was suggested by the familial aggregation of cases and the association of gene polymorphisms with CCC development. We here investigate the role of gene polymorphisms (SNPs) in several genes involved in the control of IFN-γ production and Th1 T cell differentiation in CCC development. Methods: We studied a Brazilian population including 315 CCC cases and 118 ASY subjects. We assessed 35 Tag SNPs designed to represent all the genetic information contained in the IL12B, IL10, IFNG, and IL4 genes. Results: We found 2 IL12 SNPs (rs2546893, rs919766) and a trend of association for a IL10 SNP (rs3024496) to be significantly associated with the ASY group. these associations were confirmed by multivariate analysis and allele tests. The rs919766C, 12rs2546893G, and rs3024496C alleles were associated to an increase risk to CCC development. Conclusions: Our data show that novel polymorphisms affecting IL12B and IL10, but not IFNG or IL4 genes play a role in genetic susceptibility to CCC development. This might indicate that the increased Th1 differentiation and IFN-γ production associated with CCC is genetically controlled.
Collapse
Affiliation(s)
- Amanda Farage Frade-Barros
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Aix-Marseille Université, INSERM, GIMP UMR_S906, Marseille, France.,Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, Brazil.,Bioengineering Program, Instituto Tecnológico, Universidade Brasil, São Paulo, Brazil
| | - Barbara Maria Ianni
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | | | - Cristina Wide Pissetti
- Laboratory of Immunology, Universidade Federal Do Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Bruno Saba
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Hui Tzu Lin-Wang
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Paula Buck
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - José Antonio Marin-Neto
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - André Schmidt
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Fabrício Dias
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Marcelo Sampaio
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Abílio Fragata
- Laboratório de Investigação Molecular em Cardiologia, Instituto de Cardiologia Dante Pazzanese (IDPC), São Paulo, Brazil
| | - Alexandre Costa Pereira
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil
| | - Eduardo Donadi
- School of Medicine of Ribeirão Preto (FMRP), University of São Paulo, Ribeirão Preto, Brazil
| | - Virmondes Rodrigues
- Laboratory of Immunology, Universidade Federal Do Triângulo Mineiro (UFTM), Uberaba, Brazil
| | - Jorge Kalil
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Bioengineering Program, Instituto Tecnológico, Universidade Brasil, São Paulo, Brazil
| | - Christophe Chevillard
- Aix Marseille Université, INSERM, TAGC Theories and Approaches of Genomic Complexity, UMR_1090, Marseille, France
| | - Edecio Cunha-Neto
- Heart Institute (InCor), University of São Paulo School of Medicine (FMUSP), São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, Brazil
| |
Collapse
|
17
|
Martinez SJ, Romano PS, Engman DM. Precision Health for Chagas Disease: Integrating Parasite and Host Factors to Predict Outcome of Infection and Response to Therapy. Front Cell Infect Microbiol 2020; 10:210. [PMID: 32457849 PMCID: PMC7225773 DOI: 10.3389/fcimb.2020.00210] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/16/2020] [Indexed: 01/01/2023] Open
Abstract
Chagas disease, caused by the infection with the protozoan parasite Trypanosoma cruzi, is clinically manifested in approximately one-third of infected people by inflammatory heart disease (cardiomyopathy) and, to a minor degree, gastrointestinal tract disorders (megaesophagus or megacolon). Chagas disease is a zoonosis transmitted among animals and people through the contact with triatomine bugs, which are found in much of the western hemisphere, including most countries of North, Central and South America, between parallels 45° north (Minneapolis, USA) and south (Chubut Province, Argentina). Despite much research on drug discovery for T. cruzi, there remain only two related agents in widespread use. Likewise, treatment is not always indicated due to the serious side effects of these drugs. On the other hand, the epidemiology and pathogenesis of Chagas disease are both highly complex, and much is known about both. However, it is still impossible to predict what will happen in an individual person infected with T. cruzi, because of the highly variability of parasite virulence and human susceptibility to infection, with no definitive molecular predictors of outcome from either side of the host-parasite equation. In this Minireview we briefly discuss the current state of T. cruzi infection and prognosis and look forward to the day when it will be possible to employ precision health to predict disease outcome and determine whether and when treatment of infection may be necessary.
Collapse
Affiliation(s)
- Santiago J Martinez
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora-Instituto de Histología y Embriología "Dr. Mario H. Burgos," (IHEM-CONICET- Universidad Nacional de Cuyo), Mendoza, Argentina.,Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States
| | - Patricia S Romano
- Laboratorio de Biología de Trypanosoma cruzi y la célula hospedadora-Instituto de Histología y Embriología "Dr. Mario H. Burgos," (IHEM-CONICET- Universidad Nacional de Cuyo), Mendoza, Argentina
| | - David M Engman
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, Los Angeles, CA, United States.,Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Departments of Pathology and Microbiology-Immunology, Northwestern University, Chicago, IL, United States
| |
Collapse
|
18
|
Pereira AV, Gois MB, Silva MS, Miranda Junior NRD, Campos CBHF, Schneider LCL, Barbosa CP, Nogueira-Melo GDA, Sant'Ana DDMG. Toxoplasma gondii causes lipofuscinosis, collagenopathy and spleen and white pulp atrophy during the acute phase of infection. Pathog Dis 2020; 77:5739919. [PMID: 32068829 DOI: 10.1093/femspd/ftaa008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
In this study, we evaluated homeostatic and functional disorders of the spleen in mice inoculated with Toxoplasma gondii. The kinetics of megakaryocyte and leukocyte production, body and spleen mass and certain histopathological aspects were analyzed. There was increased (P < 0.05) the accumulation of lipofuscin in the red pulp of the spleen, in the periods of 30 and 60 dpi of the infection, that is, in the chronification stage of the disease and decrease of the white pulp area. In addition, we observed (from 7dpi) a quantitative and qualitative increase (P < 0.05) in the deposition of collagen fibers in the spleen of all infected mice. Since resolution of the inflammatory process resulted in pathophysiological changes, we can suggest that the T. gondii invaded and multiplied in the cells of the white and red pulps of the spleen. Although we did not find the parasite in the spleen, this hypothesis is supported by the presence of diffuse inflammatory infiltrate, which extended through the spleen parenchyma of all inoculated mice. Taken together, our results suggest that T. gondii causes severe homeostatic disorders that have altered spleen physiology, including diffuse parenchymal inflammation, lipofuscinosis in histiocytes, early aging, collagenopathy, systemic sclerosis and spleen and white pulp atrophy.
Collapse
Affiliation(s)
- Andréia Vieira Pereira
- State University of Maringá, Department of Clinical Analysis and Biomedicine, Avenue Colombo, 5790, CEP: 87020-900, Maringá, Brazil
| | - Marcelo Biondaro Gois
- Federal University of "Recôncavo'' of Bahia, Avenue Carlos Amaral, Santo Antônio de Jesus, CEP 44.430-622, Brazil; Institute of Health Sciences, Federal University of Bahia; and Postgraduate Program in Regional Development and Environment, Maria Milza College
| | - Mariana Sacchi Silva
- State University of Maringá, Department of Clinical Analysis and Biomedicine, Avenue Colombo, 5790, CEP: 87020-900, Maringá, Brazil
| | | | - Carla Betânia Huf Ferraz Campos
- State University of Maringá, Department of Clinical Analysis and Biomedicine, Avenue Colombo, 5790, CEP: 87020-900, Maringá, Brazil
| | - Larissa Carla Lauer Schneider
- State University of Maringá, Department of Morphological Sciences, Avenue Colombo, 5790, CEP: 87020-900, Maringá, Brazil
| | - Carmem Patrícia Barbosa
- State University of Maringá, Department of Morphological Sciences, Avenue Colombo, 5790, CEP: 87020-900, Maringá, Brazil
| | | | - Débora de Mello Gonçales Sant'Ana
- State University of Maringá, Department of Clinical Analysis and Biomedicine, Avenue Colombo, 5790, CEP: 87020-900, Maringá, Brazil.,State University of Maringá, Department of Morphological Sciences, Avenue Colombo, 5790, CEP: 87020-900, Maringá, Brazil
| |
Collapse
|
19
|
Solaymani-Mohammadi S, Eckmann L, Singer SM. Interleukin (IL)-21 in Inflammation and Immunity During Parasitic Diseases. Front Cell Infect Microbiol 2019; 9:401. [PMID: 31867283 PMCID: PMC6904299 DOI: 10.3389/fcimb.2019.00401] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/11/2019] [Indexed: 12/30/2022] Open
Abstract
Parasitic diseases cause significant morbidity and mortality in the developing and underdeveloped countries. No efficacious vaccines are available against most parasitic diseases and there is a critical need for developing novel vaccine strategies for care. IL-21 is a pleiotropic cytokine whose functions in protection and immunopathology during parasitic diseases have been explored in limited ways. IL-21 and its cognate receptor, IL-21R, are highly expressed in parasitized organs of infected humans as well in murine models of the human parasitic diseases. Prior studies have indicated the ability of the IL-21/IL-21R signaling axis to regulate the effector functions (e.g., cytokine production) of T cell subsets by enhancing the expression of T-bet and STAT4 in human T cells, resulting in an augmented production of IFN-γ. Mice deficient for either IL-21 (Il21−/−) or IL-21R (Il21r−/−) showed significantly reduced inflammatory responses following parasitic infections as compared with their WT counterparts. Targeting the IL-21/IL-21R signaling axis may provide a novel approach for the development of new therapeutic agents for the prevention of parasite-induced immunopathology and tissue destruction.
Collapse
Affiliation(s)
- Shahram Solaymani-Mohammadi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Lars Eckmann
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Steven M Singer
- Department of Biology, Georgetown University, Washington, DC, United States
| |
Collapse
|
20
|
Lopes CD, Possato B, Gaspari APS, Oliveira RJ, Abram U, Almeida JPA, Rocho FDR, Leitão A, Montanari CA, Maia PIS, da Silva JS, de Albuquerque S, Carneiro ZA. Organometallic Gold(III) Complex [Au(Hdamp)(L1 4)] + (L1 = SNS-Donating Thiosemicarbazone) as a Candidate to New Formulations against Chagas Disease. ACS Infect Dis 2019; 5:1698-1707. [PMID: 31419384 DOI: 10.1021/acsinfecdis.8b00284] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chagas disease remains a serious public health concern with unsatisfactory treatment outcomes due to strain-specific drug resistance and various side effects. To identify new therapeutic drugs against Trypanosoma cruzi, we evaluated both the in vitro and in vivo activity of the organometallic gold(III) complex [Au(III)(Hdamp)(L14)]Cl (L1 = SNS-donating thiosemicarbazone), henceforth denoted 4-Cl. Our results demonstrated that 4-Cl was more effective than benznidazole (Bz) in eliminating both the extracellular trypomastigote and intracellular amastigote forms of the parasite without cytotoxic effects on mammalian cells. In in vivo assays, 4-Cl in PBS solution loses the protonation and becomes the 4-neutral. 4-Neutral reduced parasitaemia and tissue parasitism in addition to protecting the liver and heart from tissue damage at 2.8 mg/kg/day. All these changes resulted in the survival of 100% of the mice treated with the gold complex during the acute phase. Analyzing the surviving animals of the acute infection, the parasite load after 150 days of infection was equivalent to those treated with the standard dose of Bz without demonstrating the hepatotoxicity of the latter. In addition, we identified a modulation of interferon gamma (IFN-γ) levels that may be targeting the disease's positive outcome. To the best of our knowledge, this is the first gold organometallic study that shows promise in an in vivo experimental model against Chagas disease.
Collapse
Affiliation(s)
- Carla Duque Lopes
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
- Departament of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Bruna Possato
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Ana Paula S. Gaspari
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Ronaldo J. Oliveira
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais 38025-470, Brazil
| | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin D-14195, Germany
| | - José P. A. Almeida
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Fernanda dos Reis Rocho
- Grupo de Estudos em Química Medicinal de Produtos Naturais−NEQUIMED-PN, Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador Sancarlense 400, P.O. Box 780, São Carlos, São Paulo 13560-960, Brazil
| | - Andrei Leitão
- Grupo de Estudos em Química Medicinal de Produtos Naturais−NEQUIMED-PN, Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador Sancarlense 400, P.O. Box 780, São Carlos, São Paulo 13560-960, Brazil
| | - Carlos A. Montanari
- Grupo de Estudos em Química Medicinal de Produtos Naturais−NEQUIMED-PN, Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador Sancarlense 400, P.O. Box 780, São Carlos, São Paulo 13560-960, Brazil
| | - Pedro I. S. Maia
- Núcleo de Desenvolvimento de Compostos Bioativos (NDCBio), Universidade Federal do Triângulo Mineiro, Uberaba, Minas Gerais 38025-470, Brazil
| | - João S. da Silva
- Departament of Biochemistry and Immunology, School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Sérgio de Albuquerque
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Zumira A. Carneiro
- Department of Clinical Toxicological and Bromatological Analysis School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| |
Collapse
|
21
|
Does Prolactin treatment trigger imunoendocrine alterations during experimental T. cruzi infection? Cytokine 2019; 121:154736. [PMID: 31163343 DOI: 10.1016/j.cyto.2019.154736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 05/08/2019] [Accepted: 05/24/2019] [Indexed: 12/12/2022]
Abstract
Prolactin (PRL) is a pleiotropic polypeptide hormone produced by the anterior pituitary gland and negatively controlled by dopamine. Some researchers have associated the immune regulatory functions of PRL with some infectious diseases like Toxoplasma gondii and T. cruzi. This work aimed to analyze the possible immuno-modulatory effects of this hormone through the subcutaneous administration of PRL during the experimental Chagas disease. On the 14th day post-infection (dpi), PRL triggered increased percentages of NK cells in treated infected animals as compared to the infected and untreated ones. For early and late apoptosis, our results showed that in chronically infected groups, PRL counteracted splenocyte apoptosis as revealed by the reduced percentages of both, early and late apoptosis. Reduced percentages of spleen CD4+ and CD8+ T cells were detected in infected PRL treated rats (60 days post-infection). Concerning to B cells, a significant increased percentage of these cells was found for all PRL treated infected animals (14th dpi), but no statistically significant alteration was observed on the 60th days post-infection. Furthermore, PRL treatment triggered a significant increase in the percentage of CD4+ T lymphocytes IFN-γ producers, while on the 60th dpi, a reduced percentage of IFN-γ in these cells was observed in prolactin-treated rats compared to infected and untreated ones. Enhanced serum IL-12 levels were detected in infected and PRL treated subjects (60th dpi). Only on 7th day post-infection, the flow cytometric analysis of CFSE-stained CD3+ T cells showed an enhanced proliferation of polyclonal stimulated T cells in PRL-treated and infected animals. In this study, we demonstrated that PRL can influence many aspects of the immune response during the experimental Chagas' disease, and this substance could be used as a supporting trial along with the conventional drug treatment.
Collapse
|
22
|
Błyszczuk P. Myocarditis in Humans and in Experimental Animal Models. Front Cardiovasc Med 2019; 6:64. [PMID: 31157241 PMCID: PMC6532015 DOI: 10.3389/fcvm.2019.00064] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/30/2019] [Indexed: 12/21/2022] Open
Abstract
Myocarditis is defined as an inflammation of the cardiac muscle. In humans, various infectious and non-infectious triggers induce myocarditis with a broad spectrum of histological presentations and clinical symptoms of the disease. Myocarditis often resolves spontaneously, but some patients develop heart failure and require organ transplantation. The need to understand cellular and molecular mechanisms of inflammatory heart diseases led to the development of mouse models for experimental myocarditis. It has been shown that pathogenic agents inducing myocarditis in humans can often trigger the disease in mice. Due to multiple etiologies of inflammatory heart diseases in humans, a number of different experimental approaches have been developed to induce myocarditis in mice. Accordingly, experimental myocarditis in mice can be induced by infection with cardiotropic agents, such as coxsackievirus B3 and protozoan parasite Trypanosoma cruzi or by activating autoimmune responses against heart-specific antigens. In certain models, myocarditis is followed by the phenotype of dilated cardiomyopathy and the end stage of heart failure. This review describes the most commonly used mouse models of experimental myocarditis with a focus on the role of the innate and adaptive immune systems in induction and progression of the disease. The review discusses also advantages and limitations of individual mouse models in the context of the clinical manifestation and the course of the disease in humans. Finally, animal-free alternatives in myocarditis research are outlined.
Collapse
Affiliation(s)
- Przemysław Błyszczuk
- Department of Clinical Immunology, Jagiellonian University Medical College, Cracow, Poland.,Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Infection by Trypanosoma cruzi in the central nervous system in non-human mammals: a systematic review. Parasitology 2019; 146:983-1005. [PMID: 30873928 DOI: 10.1017/s0031182019000210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Currently, the types and distribution of the lesions induced in the central nervous system (CNS) by Trypanosoma cruzi remain unclear as the available evidence is based on fragmented data. Therefore, we developed a systematic review to analyse the main characteristics of the CNS lesions in non-human hosts infected. From a structured search on the PubMed/Medline and Scopus platforms, 32 studies were retrieved, subjected to data extraction and methodological bias analysis. Our results show that the most frequent alterations in the CNS are the presence of different forms of T. cruzi and intense lymphocytes infiltrates. The encephalon is the main target of T. cruzi, and inflammatory changes in the CNS are more frequent and severe in the acute phase of infection. The parasite's genotype and phenotype are associated with the tropism and severity of the CNS lesions. The methodological limitations found in the studies were divergences in inoculation pathways, under-reporting of animal age and weight, sample calculation strategies and histopathological characterization. Since the changes were dependent on the pathogenicity and virulence of the T. cruzi strains, the genotype and phenotype characterization of the parasite are extremely relevant to predict changes in the CNS and the neurological manifestations associated with Chagas' disease.
Collapse
|
24
|
Piacenza L, Trujillo M, Radi R. Reactive species and pathogen antioxidant networks during phagocytosis. J Exp Med 2019; 216:501-516. [PMID: 30792185 PMCID: PMC6400530 DOI: 10.1084/jem.20181886] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/04/2019] [Accepted: 02/04/2019] [Indexed: 11/23/2022] Open
Abstract
The generation of phagosomal cytotoxic reactive species (i.e., free radicals and oxidants) by activated macrophages and neutrophils is a crucial process for the control of intracellular pathogens. The chemical nature of these species, the reactions they are involved in, and the subsequent effects are multifaceted and depend on several host- and pathogen-derived factors that influence their production rates and catabolism inside the phagosome. Pathogens rely on an intricate and synergistic antioxidant armamentarium that ensures their own survival by detoxifying reactive species. In this review, we discuss the generation, kinetics, and toxicity of reactive species generated in phagocytes, with a focus on the response of macrophages to internalized pathogens and concentrating on Mycobacterium tuberculosis and Trypanosoma cruzi as examples of bacterial and parasitic infection, respectively. The ability of pathogens to deal with host-derived reactive species largely depends on the competence of their antioxidant networks at the onset of invasion, which in turn can tilt the balance toward pathogen survival, proliferation, and virulence over redox-dependent control of infection.
Collapse
Affiliation(s)
- Lucía Piacenza
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Rafael Radi
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
25
|
Chevillard C, Nunes JPS, Frade AF, Almeida RR, Pandey RP, Nascimento MS, Kalil J, Cunha-Neto E. Disease Tolerance and Pathogen Resistance Genes May Underlie Trypanosoma cruzi Persistence and Differential Progression to Chagas Disease Cardiomyopathy. Front Immunol 2018; 9:2791. [PMID: 30559742 PMCID: PMC6286977 DOI: 10.3389/fimmu.2018.02791] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 11/13/2018] [Indexed: 01/01/2023] Open
Abstract
Chagas disease is caused by infection with the protozoan Trypanosoma cruzi and affects over 8 million people worldwide. In spite of a powerful innate and adaptive immune response in acute infection, the parasite evades eradication, leading to a chronic persistent infection with low parasitism. Chronically infected subjects display differential patterns of disease progression. While 30% develop chronic Chagas disease cardiomyopathy (CCC)—a severe inflammatory dilated cardiomyopathy—decades after infection, 60% of the patients remain disease-free, in the asymptomatic/indeterminate (ASY) form, and 10% develop gastrointestinal disease. Infection of genetically deficient mice provided a map of genes relevant for resistance to T. cruzi infection, leading to the identification of multiple genes linked to survival to infection. These include pathogen resistance genes (PRG) needed for intracellular parasite destruction, and genes involved in disease tolerance (protection against tissue damage and acute phase death—DTG). All identified DTGs were found to directly or indirectly inhibit IFN-γ production or Th1 differentiation. We hypothesize that the absolute need for DTG to control potentially lethal IFN-γ PRG activity leads to T. cruzi persistence and establishment of chronic infection. IFN-γ production is higher in CCC than ASY patients, and is the most highly expressed cytokine in CCC hearts. Key DTGs that downmodulate IFN-γ, like IL-10, and Ebi3/IL27p28, are higher in ASY patients. Polymorphisms in PRG and DTG are associated with differential disease progression. We thus hypothesize that ASY patients are disease tolerant, while an imbalance of DTG and IFN-γ PRG activity leads to the inflammatory heart damage of CCC.
Collapse
Affiliation(s)
| | - João Paulo Silva Nunes
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Amanda Farage Frade
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.,Department of Bioengineering, Brazil University, São Paulo, Brazil
| | - Rafael Ribeiro Almeida
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Ramendra Pati Pandey
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Marilda Savóia Nascimento
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Jorge Kalil
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratorio de Imunologia, Instituto do Coracao, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Disciplina de Imunologia Clínica e Alergia, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| |
Collapse
|
26
|
Felizardo AA, Caldas IS, Mendonça AAS, Gonçalves RV, Tana FL, Almeida LA, Novaes RD. Impact of Trypanosoma cruzi infection on nitric oxide synthase and arginase expression and activity in young and elderly mice. Free Radic Biol Med 2018; 129:227-236. [PMID: 30248443 DOI: 10.1016/j.freeradbiomed.2018.09.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/21/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022]
Abstract
Elderly organisms are more susceptible to infectious diseases. However, the impact of aging on antiparasitic mechanisms, especially the nitric oxide pathway, is poorly understood. Using an integrated in vivo and in vitro model, we compared the severity of Trypanosoma cruzi infection in young and elderly (8 or 72 weeks old) mice. Forty C57BL/6 mice were randomized into four groups: Y-inf, young infected; Yn-inf, young uninfected; A-inf, aged infected; An-inf, aged uninfected. Parasitemia was measured daily, and animals were euthanized after 15 days of infection. Trypanosoma cruzi-induced inflammatory processes were analyzed in blood and heart samples, as well as in bone marrow-derived macrophages (BMDMs) co-cultured with splenocytes isolated from young or elderly mice. Our results indicated upregulated IgG2b and IL-17 production in elderly animals, which was not sufficient to reduce parasitemia, parasitic load and myocarditis to levels observed in young animals. The higher susceptibility of elderly mice to T. cruzi infection was accompanied by reduced cardiac inducible nitric oxide synthase (iNOS) gene expression, nitric oxide (NO) and IFN-γ levels, as well as an antagonistic upregulation of arginase-1 expression and arginase activity. The same responses were observed when BMDMs co-cultured with splenocytes from elderly mice were stimulated with T. cruzi antigens. Our findings indicate that elderly mice were more susceptible to T. cruzi infection, which was potentially related to an attenuated response to antigenic stimulation, inhibition of iNOS gene expression and NO production, and antagonistic upregulation of arginase gene expression and activity, which created favorable conditions for heart parasitism and myocarditis development.
Collapse
Affiliation(s)
- Amanda A Felizardo
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Ivo S Caldas
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Andréa A S Mendonça
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-000, Minas Gerais, Brazil
| | - Fernanda L Tana
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Leonardo A Almeida
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Microbiology and Immunology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil
| | - Rômulo D Novaes
- Institute of Biomedical Sciences, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil; Department of Structural Biology, Federal University of Alfenas, Alfenas, 37130-001 Minas Gerais, Brazil.
| |
Collapse
|
27
|
Upregulation of Cardiac IL-10 and Downregulation of IFN- γ in Balb/c IL-4 -/- in Acute Chagasic Myocarditis due to Colombian Strain of Trypanosoma cruzi. Mediators Inflamm 2018; 2018:3421897. [PMID: 30622430 PMCID: PMC6304210 DOI: 10.1155/2018/3421897] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
Inflammatory response in Chagas disease is related to parasite and host factors. However, immune system regulation has not been fully elucidated. Thus, this study is aimed at evaluating IL-4 influence on acute phase of Trypanosoma cruzi experimental infection through dosage of cytokine levels in cardiac homogenate of infected Balb/c WT and Balb/c IL-4−/− as well as its histopathological repercussions. For such purpose, mice were divided into two groups: an infected group with 100 forms of the Colombian strain and an uninfected group. After 21 days of infection, animals were euthanized and the blood, spleen, and heart were collected. The spleen was used to culture splenic cells in 48 h. Subsequently, cytokines TNF-α, IL-12p70, IL-10, IFN-γ, and IL-17 were measured in the blood, culture supernatant, and heart apex by ELISA. The base of the heart was used for histopathological analysis. From these analysis, infected Balb/c IL-4−/− mice showed milder inflammatory infiltrate compared to Balb/c WT, but without changes in nest density and collagen deposition. IL-4 absence culminated in lower cardiac tissue IFN-γ production, although it did not affect TNF-α expression in situ. It also decreased TNF-α systemic production and increased IL-10, both systemically and in situ. In addition, IL-4 absence did not influence IL-17 expression. Splenocytes of IL-4-deficient mice produced higher amounts of IFN-γ, TNF-α, and IL-17 and lower amounts of IL-10. Thus, IL-4 absence in acute phase of experimental infection with T. cruzi Colombian strain reduces myocarditis due to lower IFN-γ production and greater IL-10 production in situ and this pattern is not influenced by splenocyte general repertoire.
Collapse
|
28
|
Acevedo GR, Girard MC, Gómez KA. The Unsolved Jigsaw Puzzle of the Immune Response in Chagas Disease. Front Immunol 2018; 9:1929. [PMID: 30197647 PMCID: PMC6117404 DOI: 10.3389/fimmu.2018.01929] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Trypanosoma cruzi interacts with the different arms of the innate and adaptive host's immune response in a very complex and flowery manner. The history of host-parasite co-evolution has provided this protozoan with means of resisting, escaping or subverting the mechanisms of immunity and establishing a chronic infection. Despite many decades of research on the subject, the infection remains incurable, and the factors that steer chronic Chagas disease from an asymptomatic state to clinical onset are still unclear. As the relationship between T. cruzi and the host immune system is intricate, so is the amount and diversity of scientific knowledge on the matter. Many of the mechanisms of immunity are fairly well understood, but unveiling the factors that lead each of these to success or failure, within the coordinated response as a whole, requires further research. The intention behind this Review is to compile the available information on the different aspects of the immune response, with an emphasis on those phenomena that have been studied and confirmed in the human host. For ease of comprehension, it has been subdivided in sections that cover the main humoral and cell-mediated components involved therein. However, we also intend to underline that these elements are not independent, but function intimately and concertedly. Here, we summarize years of investigation carried out to unravel the puzzling interplay between the host and the parasite.
Collapse
Affiliation(s)
| | | | - Karina A. Gómez
- Laboratorio de Inmunología de las Infecciones por Tripanosomátidos, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
29
|
Novel Strategy To Adapt Simian-Human Immunodeficiency Virus E1 Carrying env from an RV144 Volunteer to Rhesus Macaques: Coreceptor Switch and Final Recovery of a Pathogenic Virus with Exclusive R5 Tropism. J Virol 2018; 92:JVI.02222-17. [PMID: 29743361 DOI: 10.1128/jvi.02222-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/17/2018] [Indexed: 02/06/2023] Open
Abstract
The phase III RV144 human immunodeficiency virus (HIV) vaccine trial conducted in Thailand remains the only study to show efficacy in decreasing the HIV acquisition risk. In Thailand, circulating recombinant forms of HIV clade A/E (CRF01_AE) predominate; in such viruses, env originates from clade E (HIV-E). We constructed a simian-human immunodeficiency virus (SHIV) chimera carrying env isolated from an RV144 placebo recipient in the SHIV-1157ipd3N4 backbone. The latter contains long terminal repeats (LTRs) with duplicated NF-κB sites, thus resembling HIV LTRs. We devised a novel strategy to adapt the parental infectious molecular clone (IMC), R5 SHIV-E1, to rhesus macaques: the simultaneous depletion of B and CD8+ cells followed by the intramuscular inoculation of proviral DNA and repeated administrations of cell-free virus. High-level viremia and CD4+ T-cell depletion ensued. Passage 3 virus unexpectedly caused acute, irreversible CD4+ T-cell loss; the partially adapted SHIV had become dual tropic. Virus and IMCs with exclusive R5 tropism were reisolated from earlier passages, combined, and used to complete adaptation through additional macaques. The final isolate, SHIV-E1p5, remained solely R5 tropic. It had a tier 2 neutralization phenotype, was mucosally transmissible, and was pathogenic. Deep sequencing revealed 99% Env amino acid sequence conservation; X4-only and dual-tropic strains had evolved independently from an early branch of parental SHIV-E1. To conclude, our primate model data reveal that SHIV-E1p5 recapitulates important aspects of HIV transmission and pathobiology in humans.IMPORTANCE Understanding the protective principles that lead to a safe, effective vaccine against HIV in nonhuman primate (NHP) models requires test viruses that allow the evaluation of anti-HIV envelope responses. Reduced HIV acquisition risk in RV144 has been linked to nonneutralizing IgG antibodies with a range of effector activities. Definitive experiments to decipher the mechanisms of the partial protection observed in RV144 require passive-immunization studies in NHPs with a relevant test virus. We have generated such a virus by inserting env from an RV144 placebo recipient into a SHIV backbone with HIV-like LTRs. The final SHIV-E1p5 isolate, grown in rhesus monkey peripheral blood mononuclear cells, was mucosally transmissible and pathogenic. Earlier SHIV-E passages showed a coreceptor switch, again mimicking HIV biology in humans. Thus, our series of SHIV-E strains mirrors HIV transmission and disease progression in humans. SHIV-E1p5 represents a biologically relevant tool to assess prevention strategies.
Collapse
|
30
|
Bivona AE, Sánchez Alberti A, Matos MN, Cerny N, Cardoso AC, Morales C, González G, Cazorla SI, Malchiodi EL. Trypanosoma cruzi 80 kDa prolyl oligopeptidase (Tc80) as a novel immunogen for Chagas disease vaccine. PLoS Negl Trop Dis 2018; 12:e0006384. [PMID: 29601585 PMCID: PMC5895069 DOI: 10.1371/journal.pntd.0006384] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/11/2018] [Accepted: 03/12/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chagas disease, also known as American Trypanosomiasis, is a chronic parasitic disease caused by the flagellated protozoan Trypanosoma cruzi that affects about 8 million people around the world where more than 25 million are at risk of contracting the infection. Despite of being endemic on 21 Latin-American countries, Chagas disease has become a global concern due to migratory movements. Unfortunately, available drugs for the treatment have several limitations and they are generally administered during the chronic phase of the infection, when its efficacy is considered controversial. Thus, prophylactic and/or therapeutic vaccines are emerging as interesting control alternatives. In this work, we proposed Trypanosoma cruzi 80 kDa prolyl oligopeptidase (Tc80) as a new antigen for vaccine development against Chagas disease. METHODOLOGY/PRINCIPAL FINDINGS In a murine model, we analyzed the immune response triggered by different immunization protocols based on Tc80 and evaluated their ability to confer protection against a challenge with the parasite. Immunized mice developed Tc80-specific antibodies which were able to carry out different functions such as: enzymatic inhibition, neutralization of parasite infection and complement-mediated lysis of trypomastigotes. Furthermore, vaccinated mice elicited strong cell-mediated immunity. Spleen cells from immunized mice proliferated and secreted Th1 cytokines (IL-2, IFN-γ and TNF-α) upon re-stimulation with rTc80. Moreover, we found Tc80-specific polyfunctional CD4 T cells, and cytotoxic T lymphocyte activity against one Tc80 MHC-I peptide. Immunization protocols conferred protection against a T. cruzi lethal challenge. Immunized groups showed a decreased parasitemia and higher survival rate compared with non-immunized control mice. Moreover, during the chronic phase of the infection, immunized mice presented: lower levels of myopathy-linked enzymes, parasite burden, electrocardiographic disorders and inflammatory cells. CONCLUSIONS/SIGNIFICANCE Considering that an early control of parasite burden and tissue damage might contribute to avoid the progression towards symptomatic forms of chronic Chagas disease, the efficacy of Tc80-based vaccines make this molecule a promising immunogen for a mono or multicomponent vaccine against T. cruzi infection.
Collapse
Affiliation(s)
- Augusto E. Bivona
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Ricardo A. Margni (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Andrés Sánchez Alberti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Ricardo A. Margni (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Marina N. Matos
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Ricardo A. Margni (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Natacha Cerny
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Ricardo A. Margni (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Alejandro C. Cardoso
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Ricardo A. Margni (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Celina Morales
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Germán González
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Silvia I. Cazorla
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Ricardo A. Margni (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET). Tucumán, Argentina
| | - Emilio L. Malchiodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral Ricardo A. Margni (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
31
|
Gatto M, Oliveira LRC, De Nuzzi Dias F, Araújo Júnior JP, Lima CRG, Lordelo EP, Dos Santos RM, Kurokawa CS. Benznidazole affects expression of Th1, Th17 and Treg cytokines during acute experimental Trypanosoma cruzi infection. J Venom Anim Toxins Incl Trop Dis 2017; 23:47. [PMID: 29255475 PMCID: PMC5727918 DOI: 10.1186/s40409-017-0137-4] [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: 03/29/2017] [Accepted: 11/24/2017] [Indexed: 01/11/2023] Open
Abstract
Background The present study evaluated the effect of treatment with benznidazole on mRNA expression of IFN-γ, IL-17, IL-10, TGF-β and FoxP3 in spleen and heart tissue of BALB/c mice in the acute phase of an experimental infection with Trypanosoma cruzi, strains JLP or Y. Methods The mRNA expression of cytokines and parasite load were assessed by q-PCR. Dependent groups were compared using Student's paired t-test and independent groups were compared using Student's unpaired t-test. Results Infection with the JLP or Y strains increased expression of IFN-γ in the heart and of IL-10 and IL-17 in the spleen and heart compared to uninfected animals. Treatment increased the expression of IFN-γ and decreased the expression of IL-17, IL-10, TGF- β and Foxp3 in spleen and heart tissue compared to untreated infected animals. Conclusion Benznidazole can induce Th1 profile in the initial of the acute phase. The treatment decreased the parasite load in both organs, although the number of parasites in Y-strain-infected mice remained high. The data suggest that benznidazole may modulate cytokine expression in infection and can be dependent of the strain. However, treatment was not fully effective in the infection provoked by Y strain, probably due to the characteristics of the strain itself.
Collapse
Affiliation(s)
- Mariana Gatto
- Department of Tropical Diseases, Botucatu Medical School, São Paulo State University (UNESP - Univ Estadual Paulista), Av. Professor Mário Rubens Guimarães Montenegro, s/n, Distrito de Rubião Júnior, Botucatu, 18.6186-87 SP Brazil
| | - Larissa Ragozo Cardoso Oliveira
- Department of Microbiology and Immunology, Botucatu Biosciences Institute, São Paulo State University (UNESP - Univ Estadual Paulista), Botucatu, SP Brazil
| | - Fernanda De Nuzzi Dias
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP - Univ Estadual Paulista), Araraquara, SP Brazil
| | - João Pessoa Araújo Júnior
- Department of Microbiology and Immunology, Botucatu Biosciences Institute, São Paulo State University (UNESP - Univ Estadual Paulista), Botucatu, SP Brazil
| | - Carlos Roberto Gonçalves Lima
- Department of Tropical Diseases, Botucatu Medical School, São Paulo State University (UNESP - Univ Estadual Paulista), Av. Professor Mário Rubens Guimarães Montenegro, s/n, Distrito de Rubião Júnior, Botucatu, 18.6186-87 SP Brazil
| | - Eliana Peresi Lordelo
- Department of Immunology, University of Western São Paulo (Unoeste), Presidente Prudente, SP Brazil
| | - Rodrigo Mattos Dos Santos
- Department of Tropical Diseases, Botucatu Medical School, São Paulo State University (UNESP - Univ Estadual Paulista), Av. Professor Mário Rubens Guimarães Montenegro, s/n, Distrito de Rubião Júnior, Botucatu, 18.6186-87 SP Brazil
| | - Cilmery Suemi Kurokawa
- Department of Tropical Diseases, Botucatu Medical School, São Paulo State University (UNESP - Univ Estadual Paulista), Av. Professor Mário Rubens Guimarães Montenegro, s/n, Distrito de Rubião Júnior, Botucatu, 18.6186-87 SP Brazil
| |
Collapse
|
32
|
Sandri PF, Portocarrero AR, Ciupa L, Ferraz FN, Falkowski-Temporini GJ, Rodrigues WN, Ferreira ÉC, Aleixo DL, de Araújo SM. Dynamized ethyl alcohol improves immune response and behavior in murine infection with Trypanosoma cruzi. Cytokine 2017; 99:240-248. [DOI: 10.1016/j.cyto.2017.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 12/29/2022]
|
33
|
Oda JY, Belém MO, Carlos TM, Gouveia R, Luchetti BFC, Moreira NM, Massocatto CL, Araújo SM, Sant Ana DMG, Buttow NC, Pinge-Filho P, Araújo EJA. Myenteric neuroprotective role of aspirin in acute and chronic experimental infections with Trypanosoma cruzi. Neurogastroenterol Motil 2017; 29:1-13. [PMID: 28524628 DOI: 10.1111/nmo.13102] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 04/05/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Experimental and clinical studies have shown that myenteric neuron cell death during infection with Trypanosoma cruzi mainly occurs in the esophagus and colon, resulting in megaesophagus and megacolon, respectively. Evidence suggests that the cyclooxygenase enzyme (COX) is involved in the T. cruzi invasion process. The use of low-dose aspirin (ASA), a COX-1/COX-2 inhibitor, has been shown to reduce infection with T. cruzi. Therefore, in this study, we evaluated the effects of treatment with low-dose ASA on myenteric colonic neurons during murine infection with T. cruzi. METHODS Swiss mice were assigned into groups treated with either phosphate-buffered saline or low doses of ASA during the acute phase (20 mg/kg ASA) and chronic phase (50 mg/kg ASA) of infection with the Y strain of T. cruzi. Seventy-five days after infection, colon samples were collected to quantify inflammatory foci in histological sections and also general (myosin-V+ ), nitrergic, and VIPergic myenteric neurons in whole mounts. Gastrointestinal transit time was also measured. KEY RESULTS Aspirin treatment during the acute phase of infection reduced parasitemia (P<.05). Aspirin treatment during the acute or chronic phase of the infection reduced the intensity of inflammatory foci in the colon, protected myenteric neurons from cell death and plastic changes, and recovered the gastrointestinal transit of mice infected with T. cruzi (P<.05). CONCLUSION & INFERENCES Early and delayed treatment with low-dose ASA can reduce the morphofunctional damage of colonic myenteric neurons caused by murine T. cruzi infection.
Collapse
Affiliation(s)
- J Y Oda
- Department of Medicine, Federal University of Mato Grosso do Sul, Três Lagoas, Mato Grosso do Sul, Brazil.,Department of Pathological Science, State University of Londrina, Londrina, Paraná, Brazil
| | - M O Belém
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - T M Carlos
- Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - R Gouveia
- Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| | - B F C Luchetti
- Department of Pathological Science, State University of Londrina, Londrina, Paraná, Brazil
| | - N M Moreira
- Center for Education, Letters and Health, State University of Western Paraná, Foz do Iguaçu, Paraná, Brazil
| | - C L Massocatto
- Department of Morphological Science, State University of Maringá, Maringá, Paraná, Brazil
| | - S M Araújo
- Department of Basic Health Science, State University of Maringá, Maringá, Paraná, Brazil
| | - D M G Sant Ana
- Department of Morphological Science, State University of Maringá, Maringá, Paraná, Brazil
| | - N C Buttow
- Department of Morphological Science, State University of Maringá, Maringá, Paraná, Brazil
| | - P Pinge-Filho
- Department of Pathological Science, State University of Londrina, Londrina, Paraná, Brazil
| | - E J A Araújo
- Department of Histology, State University of Londrina, Londrina, Paraná, Brazil
| |
Collapse
|
34
|
Oliveira AC, Gomes-Neto JF, Barbosa CHD, Granato A, Reis BS, Santos BM, Fucs R, Canto FB, Nakaya HI, Nóbrega A, Bellio M. Crucial role for T cell-intrinsic IL-18R-MyD88 signaling in cognate immune response to intracellular parasite infection. eLife 2017; 6:30883. [PMID: 28895840 PMCID: PMC5629024 DOI: 10.7554/elife.30883] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 09/12/2017] [Indexed: 12/29/2022] Open
Abstract
MyD88 is the main adaptor molecule for TLR and IL-1R family members. Here, we demonstrated that T-cell intrinsic MyD88 signaling is required for proliferation, protection from apoptosis and expression of activation/memory genes during infection with the intracellular parasite Trypanosoma cruzi, as evidenced by transcriptome and cytometry analyses in mixed bone-marrow (BM) chimeras. The lack of direct IL-18R signaling in T cells, but not of IL-1R, phenocopied the absence of the MyD88 pathway, indicating that IL-18R is a critical MyD88-upstream pathway involved in the establishment of the Th1 response against an in vivo infection, a presently controvert subject. Accordingly, Il18r1−/− mice display lower levels of Th1 cells and are highly susceptible to infection, but can be rescued from mortality by the adoptive transfer of WT CD4+ T cells. Our findings establish the T-cell intrinsic IL-18R/MyD88 pathway as a crucial element for induction of cognate Th1 responses against an important human pathogen.
Collapse
Affiliation(s)
- Ana-Carolina Oliveira
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Francisco Gomes-Neto
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Alessandra Granato
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Bruno Maia Santos
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rita Fucs
- Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Fábio B Canto
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Helder I Nakaya
- Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil.,Instituto Nacional de Ciência e Tecnologia de Vacinas, CNPq-MCT, Belo Horizonte, Brazil.,Department of Pathology, Emory University School of Medicine, Atlanta, United States
| | - Alberto Nóbrega
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Bellio
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia de Vacinas, CNPq-MCT, Belo Horizonte, Brazil
| |
Collapse
|
35
|
Freire-de-Lima L, Gentile LB, da Fonseca LM, da Costa KM, Santos Lemos J, Jacques LR, Morrot A, Freire-de-Lima CG, Nunes MP, Takiya CM, Previato JO, Mendonça-Previato L. Role of Inactive and Active Trypanosoma cruzi Trans-sialidases on T Cell Homing and Secretion of Inflammatory Cytokines. Front Microbiol 2017; 8:1307. [PMID: 28744279 PMCID: PMC5504189 DOI: 10.3389/fmicb.2017.01307] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/28/2017] [Indexed: 12/29/2022] Open
Abstract
Trans-sialidase from Trypanosoma cruzi (Tc-TS) belongs to a superfamily of proteins that may have enzymatic activity. While enzymatically active members (Tc-aTS) are able to transfer sialic acid from the host cell sialyl-glycoconjugates onto the parasite or to other molecules on the host cell surface, the inactive members (Tc-iTS) are characterized by their lectinic properties. Over the last 10 years, several papers demonstrated that, individually, Tc-aTS or Tc-iTS is able to modulate several biological events. Since the genes encoding Tc-iTS and Tc-aTS are present in the same copy number, and both proteins portray similar substrate-specificities as well, it would be plausible to speculate that such molecules may compete for the same sialyl-glycan structures and govern numerous immunobiological phenomena. However, their combined effect has never been evaluated in the course of an acute infection. In this study, we investigated the ability of both proteins to modulate the production of inflammatory signals, as well as the homing of T cells to the cardiac tissue of infected mice, events that usually occur during the acute phase of T. cruzi infection. The results showed that the intravenous administration of Tc-iTS, but not Tc-aTS protected the cardiac tissue from injury caused by reduced traffic of inflammatory cells. In addition, the ability of Tc-aTS to modulate the production of inflammatory cytokines was attenuated and/or compromised when Tc-iTS was co-injected in the same proportions. These results suggest that although both proteins present structural similarities and compete for the same sialyl-glycan epitopes, they might present distinct immunomodulatory properties on T cells following T. cruzi infection.
Collapse
Affiliation(s)
- Leonardo Freire-de-Lima
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Luciana B Gentile
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Leonardo M da Fonseca
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Kelli M da Costa
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Jessica Santos Lemos
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Lucas Rodrigues Jacques
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Alexandre Morrot
- Instituto Oswaldo Cruz, Fundação Oswaldo CruzRio de Janeiro, Brazil.,Instituto de Microbiologia, Centro de Ciência da Saúde - Sala D1-035, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Célio G Freire-de-Lima
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Marise P Nunes
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil.,Instituto Oswaldo Cruz, Fundação Oswaldo CruzRio de Janeiro, Brazil
| | - Christina M Takiya
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Jose O Previato
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Lucia Mendonça-Previato
- Laboratório de Glicobiologia, Instituto de Biofísica, Centro de Ciência da Saúde, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
36
|
Pereira AV, Góis MB, Lera KRJL, Falkowski-Temporini GJ, Massini PF, Drozino RN, Aleixo DL, Miranda MM, da Silva Watanabe P, Conchon-Costa I, da Costa IN, Dos Anjos Neto Filho M, de Araújo SM, Pavanelli WR. Histopathological lesions in encephalon and heart of mice infected with Toxoplasma gondii increase after Lycopodium clavatum 200dH treatment. Pathol Res Pract 2016; 213:50-57. [PMID: 27894616 DOI: 10.1016/j.prp.2016.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 10/26/2016] [Accepted: 11/03/2016] [Indexed: 02/07/2023]
Abstract
In many cases, symptoms of toxoplasmosis are mistaken for the ones of other infectious diseases. Clinical signs are rare in immunocompetent people. However, when they arise, in the acute phase of infection, several organs are affected due to the rapid spread of tachyzoites through the bloodstream. In the present study, the reduction of tachyzoites in peripheral blood of mice of G72 (infected 72h after treatment) and G48 (infected 48h after treatment and treated three more times), when compared with IC (infected and non-treated), suggests protective effect exerted by Lycopodium clavatum. If on the one hand L. clavatum brought benefits, reducing parasitemia, on the other hand, the parasitism became exacerbated. Histopathological analysis demonstrated focal, multifocal and diffuse inflammatory infiltrates, ranging from absent, discreet, moderate to intense, in heart and encephalon of mice of NIC (non-infected and non-treated), IC, G48 and G72 groups, respectively. In the perivascular region and meninges, the injuries were enlarged. The presence of tachyzoites was demonstrated through immunohistochemical (IHC) assay in myocardium. Toxoplasma gondii induced increase of collagen fibers in myocardium of mice of G72 and G48 groups, compared with IC (p<0.05) and NIC (p<0.001). The presence of inflammatory infiltrates, as well as the progressive fibrosis, caused myocardial remodeling in animals treated with L. clavatum. Counterstaining with H&E suggests TGF-β expression by mononuclear cells in the inflammatory infiltrate. Based on our results, we can conclude that the adopted regimen and potency exerted a protective effect, reducing parasitemia. However, it intensified the histopathological lesions in encephalon and heart of mice infected with T. gondii.
Collapse
Affiliation(s)
- Andréia Vieira Pereira
- Department of Experimental Pathology, State University of Londrina, Londrina, PR, Brazil
| | - Marcelo Biondaro Góis
- Department of Morphological Sciences, State University of Maringa, Maringa, PR, Brazil.
| | | | | | | | | | - Denise Lessa Aleixo
- Department of Health Sciences, State University of Maringa, Maringa, PR, Brazil
| | | | | | - Ivete Conchon-Costa
- Department of Experimental Pathology, State University of Londrina, Londrina, PR, Brazil
| | | | | | | | | |
Collapse
|
37
|
Abstract
Previous studies have emphasized the role of Toll-like receptors (TLRs) and myeloid differentiation factor 88 (MyD88) during infection with protozoan parasites. TLR2 was shown to be important for induction of cytokine synthesis by macrophages exposed to the purified glycosylphosphatidylinositol (GPI)-anchored mucin-like glycoproteins of Trypanosoma cruzi trypomastigotes (tGPIm). On the other hand, MyD88 —/— mice, but not TLR2 —/— mice, showed impaired cytokine production and resistance to infection with T. cruzi parasites. Here we evaluate the importance of MyD88 and TLR2 in MAPK activation and cytokine synthesis by macrophages exposed to live T. cruzi parasites and compared to tGPIm. The absence of MAPK phosphorylation in TLR2- and MyD88-deficient macrophages exposed to tGPIm correlated with the incapacity to induce cytokine release in these cells. In contrast, activation of MAPK and synthesis of pro-inflammatory cytokines were not abrogated in TLR2-deficient macrophages exposed to live T. cruzi parasites. We also showed that pretreatment with tGPIm significantly reduces cytokine release by macrophages in response to T. cruzi in a TLR2-dependent manner. Consistently, TLR2-/- mice were shown to produce enhanced levels of cytokines upon in vivo challenge with T. cruzi parasites. Together, these results suggest the involvement of additional TLR(s) in the pro-inflammatory response of macrophages to whole parasites, and that, in vivo, TLR2 may have a predominant immunoregulatory role during acute infection with T. cruzi parasites .
Collapse
|
38
|
Nabarro Ferraz F, da Veiga FK, Lessa Aleixo D, Ciupa L, de Abreu Filho BA, da Silva SS, Conchon-Costa I, Pavanelli WR, de Araújo SM. Biotherapies of rabbit serum modulate the immune response and decrease parasite load in mice infected with Trypanosoma cruzi. J Appl Biomed 2016. [DOI: 10.1016/j.jab.2015.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
39
|
Böhme J, Roßnagel C, Jacobs T, Behrends J, Hölscher C, Erdmann H. Epstein-Barr virus-induced gene 3 suppresses T helper type 1, type 17 and type 2 immune responses after Trypanosoma cruzi infection and inhibits parasite replication by interfering with alternative macrophage activation. Immunology 2016; 147:338-48. [PMID: 26694585 DOI: 10.1111/imm.12565] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 11/18/2015] [Accepted: 12/04/2015] [Indexed: 12/20/2022] Open
Abstract
The Epstein-Barr virus-induced gene 3 (EBI3) is a member of the interleukin-12 (IL)-12) family structurally related to the subunit p40 of IL-12 and forms a heterodimer either with the p28 subunit to build IL-27 or with p35 to form IL-35. Interleukin-27 is secreted by antigen-presenting cells whereas IL-35 appears to be produced mainly by regulatory T cells and regulatory B cells but both cytokines negatively regulate inflammatory immune responses. We here analysed the function of EBI3 during infection with the intracellular parasite Trypanosoma cruzi. Compared with C57BL/6 wild-type mice, EBI3-deficient (EBI3(-/-) ) mice showed a higher parasitaemia associated with an increased mortality rate. The EBI3(-/-) mice displayed an elevated inflammatory immune response with an increased production of T helper type 1 (Th1-), Th2- and Th17-derived cytokines. The increased Th2 immune response appears to have over-ridden the otherwise protective Th1 and Th17 immune responses by the induction of arginase-1-expressing alternatively activated macrophages in these mice. Hence, neutralization of IL-4 and arginase-1 activity partially restored protective immune responses in EBI3(-/-) mice. So far, our results demonstrate that EBI3 is an essential general regulator of inflammatory immune responses in experimental Chagas disease and is required for control of T. cruzi infection by inhibiting Th2-dependent alternative macrophage activation. Further studies are needed to dissect the underlying mechanisms and clarify whether EBI3 association with IL-27 or/and IL-35 accounts for its anti-inflammatory character in parasitic disease.
Collapse
Affiliation(s)
- Julia Böhme
- Infection Immunology, Research Centre Borstel, Borstel, Germany.,Cluster of Excellence Inflammation-at-Interfaces, Borstel-Kiel-Lübeck-Plön, Germany.,Singapore Immunology Network, Singapore, Singapore
| | | | - Thomas Jacobs
- Department of Immunology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - Jochen Behrends
- Infection Immunology, Research Centre Borstel, Borstel, Germany.,Fluorescence Cytometry Core Unit, Research Centre Borstel, Borstel, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Centre Borstel, Borstel, Germany.,Cluster of Excellence Inflammation-at-Interfaces, Borstel-Kiel-Lübeck-Plön, Germany
| | - Hanna Erdmann
- Infection Immunology, Research Centre Borstel, Borstel, Germany.,Cluster of Excellence Inflammation-at-Interfaces, Borstel-Kiel-Lübeck-Plön, Germany
| |
Collapse
|
40
|
Highly diluted medication reduces tissue parasitism and inflammation in mice infected by Trypanosoma cruzi. HOMEOPATHY 2016; 105:186-93. [DOI: 10.1016/j.homp.2015.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 05/13/2015] [Accepted: 09/21/2015] [Indexed: 11/19/2022]
|
41
|
Ersching J, Vasconcelos JR, Ferreira CP, Caetano BC, Machado AV, Bruna–Romero O, Baron MA, Ferreira LRP, Cunha-Neto E, Rock KL, Gazzinelli RT, Rodrigues MM. The Combined Deficiency of Immunoproteasome Subunits Affects Both the Magnitude and Quality of Pathogen- and Genetic Vaccination-Induced CD8+ T Cell Responses to the Human Protozoan Parasite Trypanosoma cruzi. PLoS Pathog 2016; 12:e1005593. [PMID: 27128676 PMCID: PMC4851296 DOI: 10.1371/journal.ppat.1005593] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 04/01/2016] [Indexed: 11/18/2022] Open
Abstract
The β1i, β2i and β5i immunoproteasome subunits have an important role in defining the repertoire of MHC class I-restricted epitopes. However, the impact of combined deficiency of the three immunoproteasome subunits in the development of protective immunity to intracellular pathogens has not been investigated. Here, we demonstrate that immunoproteasomes play a key role in host resistance and genetic vaccination-induced protection against the human pathogen Trypanosoma cruzi (the causative agent of Chagas disease), immunity to which is dependent on CD8+ T cells and IFN-γ (the classical immunoproteasome inducer). We observed that infection with T. cruzi triggers the transcription of immunoproteasome genes, both in mice and humans. Importantly, genetically vaccinated or T. cruzi-infected β1i, β2i and β5i triple knockout (TKO) mice presented significantly lower frequencies and numbers of splenic CD8+ effector T cells (CD8+CD44highCD62Llow) specific for the previously characterized immunodominant (VNHRFTLV) H-2Kb-restricted T. cruzi epitope. Not only the quantity, but also the quality of parasite-specific CD8+ T cell responses was altered in TKO mice. Hence, the frequency of double-positive (IFN-γ+/TNF+) or single-positive (IFN-γ+) cells specific for the H-2Kb-restricted immunodominant as well as subdominant T. cruzi epitopes were higher in WT mice, whereas TNF single-positive cells prevailed among CD8+ T cells from TKO mice. Contrasting with their WT counterparts, TKO animals were also lethally susceptible to T. cruzi challenge, even after an otherwise protective vaccination with DNA and adenoviral vectors. We conclude that the immunoproteasome subunits are key determinants in host resistance to T. cruzi infection by influencing both the magnitude and quality of CD8+ T cell responses. CD8+ t lymphocytes are cells of the immune system that mediate control of intracellular infections by viruses, prokaryote as well as eukaryote pathogens. To confer protection, these lymphocytes need to be elicited by pathogen peptides that are presented in association with MHC class I molecules. The degradation of self and microbial proteins by catalytic domains of the cytosolic proteasome β1, β2 and β5 subunits is intimately linked to the generation of MHC class I-restricted epitopes, which in turn are important determinants of the kinetics, specificity and efficiency of CD8+ T cell-mediated immunity. Importantly, inflammatory stimuli trigger the expression of the inducible alternative β1i, β2i and β5i subunits that form the immunoproteasomes. The qualitative and quantitative importance of immunoproteasomes in generating CD8+ T cell epitopes has recently been demonstrated in mice that are simultaneously devoid of the β1i, β2i and β5i subunits. In this study, we explored the role of immunoproteasomes in host resistance to Trypanosoma cruzi, a protozoan parasite that causes Chagas disease. We found that β1i, β2i and β5i triply deficient mice have an impaired response of CD8+ T cells and are highly susceptible to primary infection with T. cruzi. We also demonstrated that host resistance induced by a genetic vaccine able to protect normal mice from T. cruzi challenge fails to do so in the immunoproteasome-deficient mice. Our study provides strong evidences that β1i, β2i and β5i immunoproteasome subunits are important determinants of both the magnitude and quality of CD8+ T cell responses as well as immune-mediated host resistance to a human pathogen.
Collapse
Affiliation(s)
- Jonatan Ersching
- Centro de Terapia Celular e Molecular and Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, São Paulo, Brazil
| | - José R. Vasconcelos
- Centro de Terapia Celular e Molecular and Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, São Paulo, Brazil
- Departamento de Biociências, Universidade Federal de São Paulo, Santos, São Paulo, Brazil
| | - Camila P. Ferreira
- Centro de Terapia Celular e Molecular and Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, São Paulo, Brazil
| | - Braulia C. Caetano
- Departments of Medicine and Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | | | - Oscar Bruna–Romero
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Monique A. Baron
- Instituto do Coração (InCor), Faculdade de Medicina - Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Ludmila R. P. Ferreira
- Instituto do Coração (InCor), Faculdade de Medicina - Universidade de São Paulo, São Paulo, São Paulo, Brazil
- Universidade Santo Amaro, São Paulo, São Paulo, Brazil
| | - Edécio Cunha-Neto
- Instituto do Coração (InCor), Faculdade de Medicina - Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Kenneth L. Rock
- Departments of Medicine and Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ricardo T. Gazzinelli
- Departments of Medicine and Pathology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Centro de Pesquisas René Rachou, FIOCRUZ, Belo Horizonte, Minas Gerais, Brazil
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| | - Maurício M. Rodrigues
- Centro de Terapia Celular e Molecular and Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Nascentes GAN, Hernández CG, Rabelo RADS, Coelho RF, Morais FRD, Marques T, Batista LR, Meira WSF, Oliveira CJFD, Lages Silva E, Ramírez LE. The Driving of Immune Response by Th1 Adjuvants in Immunization of Mice with Trypanosoma cruzi marinkellei Elicits a Controversial Infection Control. Vector Borne Zoonotic Dis 2016; 16:317-25. [PMID: 26959861 DOI: 10.1089/vbz.2015.1874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In previous studies, we have demonstrated that inoculation with a Trypanosoma cruzi marinkellei (avirulent RM1 strain) was able to reduce parasitemia in mice challenged with T. cruzi, although it was not able to prevent histopathological lesions. Th1 response stimulation by immunization is necessary for T. cruzi infection control, but the resistance is also dependent on immunoregulatory mechanisms, which can be induced by adjuvants. Thus, we evaluated whether inoculation of T. cruzi marinkellei associated with administration of different adjuvants would be capable of inducing different patterns of immune response to maximize the immune response against T. cruzi (virulent Romildo strain) infection. Two hundred eighty nonisogenic mice were divided into 14 groups according to the immunization scheme and the subsequent challenge with virulent Romildo T. cruzi strain. Nonimmunized groups and animals inoculated without adjuvants were also included. Immune protection was not observed with Th2 adjuvants (incomplete Freund's adjuvant [IFA] and Alum) due to high parasitemia. Th1/Th2-polarizing adjuvants also did not induce immune protection because inulin was unable to maintain survival, and immune-stimulating complexes induced intense inflammatory processes. Animals sensitized with RM1 strain without adjuvants were able to reduce parasitemia, increase survival, and protect against severe histological lesions, followed by adequate cytokine stimulation. Finally, our results demonstrate that the early and balanced IFN-γ production becomes critical to promote protection and that Th1 adjuvant elicited a controversial infection control due to increased histopathological damage. Therefore, the host's immunomodulation remains one of the most important challenges in the research for effective protection against T. cruzi infection. Similarly, the identification of protective antigens in the RM1 strain of T. cruzi marinkellei may contribute to further studies on vaccine development against human Chagas disease.
Collapse
Affiliation(s)
- Gabriel Antonio Nogueira Nascentes
- 1 Microbiology and Immunology Discipline, Federal Institute of Education , Science and Technology at Triângulo Mineiro (IFTM), Uberaba, Brazil
| | - César Gómez Hernández
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Rosiley Aparecida de Souza Rabelo
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Raquel Fernandes Coelho
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Fabiana Rossetto de Morais
- 3 School of Pharmaceutical Sciences at Ribeirão Preto, University of São Paulo (USP) , Ribeirão Preto, Brazil
| | - Tatiane Marques
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Lara Rocha Batista
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Wendell Sérgio Ferreira Meira
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Carlo José Freire de Oliveira
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Eliane Lages Silva
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| | - Luis Eduardo Ramírez
- 2 Department of Microbiology, Immunology and Parasitology, Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro (UFTM) , Uberaba, Brazil
| |
Collapse
|
43
|
Gruendling AP, Massago M, Teston APM, Monteiro WM, Kaneshima EN, Araújo SM, Gomes ML, Barbosa MDGV, Toledo MJO. Impact of benznidazole on infection course in mice experimentally infected with Trypanosoma cruzi I, II, and IV. Am J Trop Med Hyg 2015; 92:1178-89. [PMID: 25940197 DOI: 10.4269/ajtmh.13-0690] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 11/01/2014] [Indexed: 11/07/2022] Open
Abstract
American trypanosomiasis is an emerging zoonosis in the Brazilian Amazon. Studies on benznidazole (BZ) chemotherapy with Trypanosoma cruzi from this region have great relevance, given the different discrete typing units (DTUs) that infect humans in the Amazon and other regions of Brazil. We performed a parasitological, histopathological, and molecular analysis of mice inoculated with strains of T. cruzi I, II, and IV that were BZ-treated during the acute phase of infection. Groups of Swiss mice were inoculated; 13 received oral BZ, whereas the other 13 comprised the untreated controls. Unlike parasitemia, the infectivity and mortality did not vary among the DTUs. Trypanosoma cruzi DNA was detected in all tissues analyzed and the proportion of organs parasitized varied with the parasite DTU. The BZ treatment reduced the most parasitological parameters, tissue parasitism and the inflammatory processes at all infection stages and for all DTUs. However, the number of significant reductions varied according to the DTU and infection phase.
Collapse
Affiliation(s)
- Ana Paula Gruendling
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Miyoko Massago
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Ana Paula M Teston
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Wuelton M Monteiro
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Edilson N Kaneshima
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Silvana M Araújo
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Mônica L Gomes
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Maria das Graças V Barbosa
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| | - Max Jean O Toledo
- Post-Graduate Program in Health Sciences (Programa de Pós-graduação em Ciências da Saúde), Maringá State University (Universidade Estadual de Maringá, UEM), Maringá, Paraná, Brazil; Department of Basic Health Sciences, Health Sciences Center, (Departamento de Ciências Básicas da Saúde, Centro de Ciências da Saúde), UEM, Maringá, Paraná, Brazil; Post-Graduate Program in Tropical Medicine (Programa de Pós-Graduação em Medicina Tropical), Dr. Heitor Vieira Dourado Foundation for Tropical Medicine (Fundação de Medicina Tropical Dr. Heitor Vieira Dourado)/Amazonas State University (Universidade do Estado do Amazonas), Manaus, Amazonas, Brazil
| |
Collapse
|
44
|
Zamboni DS, Lima-Junior DS. Inflammasomes in host response to protozoan parasites. Immunol Rev 2015; 265:156-71. [DOI: 10.1111/imr.12291] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Dario S. Zamboni
- Department of Cell Biology; School of Medicine of Ribeirão Preto; University of São Paulo; Ribeirão Preto Brazil
| | - Djalma S. Lima-Junior
- Department of Cell Biology; School of Medicine of Ribeirão Preto; University of São Paulo; Ribeirão Preto Brazil
| |
Collapse
|
45
|
Abstract
Parasitic diseases are a serious global health concern. Many of the most common and most severe parasitic diseases, including Chagas' disease, leishmaniasis, and schistosomiasis, are also classified as neglected tropical diseases and are comparatively less studied than infectious diseases prevalent in high income nations. The NLRs (nucleotide-binding domain leucine-rich-repeat-containing proteins) are cytosolic proteins known to be involved in pathogen detection and host response. The role of NLRs in the host response to parasitic infection is just beginning to be understood. The NLR proteins NOD1 and NOD2 have been shown to contribute to immune responses during Trypanosoma cruzi infection, Toxoplasma gondii infection, and murine cerebral malaria. The NLRP3 inflammasome is activated by T. cruzi and Leishmania amazonensis but also induces pathology during infection with schistosomes or malaria. Both the NLRP1 and NLRP3 inflammasomes respond to T. gondii infection. The NLRs may play crucial roles in human immune responses during parasitic infection, usually acting as innate immune sensors and driving the inflammatory response against invading parasites. However, this inflammatory response can either kill the invading parasite or be responsible for destructive pathology. Therefore, understanding the role of the NLR proteins will be critical to understanding the host defense against parasites as well as the fine balance between homeostasis and parasitic disease.
Collapse
Affiliation(s)
- Gwendolyn M Clay
- The Interdisciplinary Program in Molecular and Cellular Biology, University of Iowa, 400 EMRB, 500 Newton Rd., Iowa City, IA, 52242, USA
| | | | | |
Collapse
|
46
|
Silva RR, Mariante RM, Silva AA, dos Santos ALB, Roffê E, Santiago H, Gazzinelli RT, Lannes-Vieira J. Interferon-gamma promotes infection of astrocytes by Trypanosoma cruzi. PLoS One 2015; 10:e0118600. [PMID: 25695249 PMCID: PMC4335051 DOI: 10.1371/journal.pone.0118600] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 01/09/2015] [Indexed: 12/20/2022] Open
Abstract
The inflammatory cytokine interferon-gamma (IFNγ) is crucial for immunity against intracellular pathogens such as the protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease (CD). IFNγ is a pleiotropic cytokine which regulates activation of immune and non-immune cells; however, the effect of IFNγ in the central nervous system (CNS) and astrocytes during CD is unknown. Here we show that parasite persists in the CNS of C3H/He mice chronically infected with the Colombian T. cruzi strain despite the increased expression of IFNγ mRNA. Furthermore, most of the T. cruzi-bearing cells were astrocytes located near IFNγ+ cells. Surprisingly, in vitro experiments revealed that pretreatment with IFNγ promoted the infection of astrocytes by T. cruzi increasing uptake and proliferation of intracellular forms, despite inducing increased production of nitric oxide (NO). Importantly, the effect of IFNγ on T. cruzi uptake and growth is completely blocked by the anti-tumor necrosis factor (TNF) antibody Infliximab and partially blocked by the inhibitor of nitric oxide synthesis L-NAME. These data support that IFNγ fuels astrocyte infection by T. cruzi and critically implicate IFNγ-stimulated T. cruzi-infected astrocytes as sources of TNF and NO, which may contribute to parasite persistence and CNS pathology in CD.
Collapse
Affiliation(s)
- Rafael Rodrigues Silva
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro, Brasil
| | - Rafael M. Mariante
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro, Brasil
| | - Andrea Alice Silva
- Laboratório Multidisciplinar de Apoio à Pesquisa, Departamento de Medicina Clínica, Universidade Federal Fluminense, Rio de Janeiro, Brasil
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Rio de Janeiro, Brasil
| | | | - Ester Roffê
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro, Brasil
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisas René Rachou—Fiocruz, Minas Gerais, Brasil
| | - Helton Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Ricardo Tostes Gazzinelli
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Minas Gerais, Brasil
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações, Instituto Oswaldo Cruz—Fiocruz, Rio de Janeiro, Brasil
| |
Collapse
|
47
|
Stahl P, Schwarz RT, Debierre-Grockiego F, Meyer T. Trypanosoma cruzi parasites fight for control of the JAK-STAT pathway by disarming their host. JAKSTAT 2015; 3:e1012964. [PMID: 26413423 DOI: 10.1080/21623996.2015.1012964] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 12/13/2022] Open
Abstract
The zoonotic Chagas' disease is caused by infections with the hemoflagellate Trypanosoma cruzi (T. cruzi) which is endemic in Latin America. Despite recent advances in our understanding of the pathogenesis of the disease, the underlying molecular processes involved in host-parasite interactions are only poorly understood. In particular, the mechanisms for parasite persistence in host cells remain largely unknown. Cytokine-driven transcription factors from the family of STAT (signal transducer and activator of transcription) proteins appear to play a central role in the fight against T. cruzi infection. However, amastigotes proliferating in the cytoplasm of infected host cells develop effective strategies to circumvent the attack executed by STAT proteins. This review highlights the interactions between T. cruzi parasites and human host cells in terms of cytokine signaling and, in particular, discusses the impact of STATs on the balance between parasite invasion and clearance.
Collapse
Affiliation(s)
- Philipp Stahl
- Institute of Virology; Parasitology Unit; University of Marburg ; Marburg, Germany
| | - Ralph T Schwarz
- Institute of Virology; Parasitology Unit; University of Marburg ; Marburg, Germany ; Laboratory for Structural and Functional Glycobiology; University of Lille 1 for Sciences and Technologies ; Lille, France
| | - Françoise Debierre-Grockiego
- Mixed Research Unit 1282; François Rabelais University of Tours-INRA; Infectious Diseases and Public Health ; Tours, France
| | - Thomas Meyer
- Department of Psychosomatic Medicine and Psychotherapy; University of Göttingen ; Göttingen, Germany ; German Center for Cardiovascular Research ; Göttingen, Germany
| |
Collapse
|
48
|
Stahl P, Ruppert V, Schwarz RT, Meyer T. Trypanosoma cruzi evades the protective role of interferon-gamma-signaling in parasite-infected cells. PLoS One 2014; 9:e110512. [PMID: 25340519 PMCID: PMC4207753 DOI: 10.1371/journal.pone.0110512] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/23/2014] [Indexed: 12/15/2022] Open
Abstract
The protozoan parasite Trypanosoma cruzi is responsible for the zoonotic Chagas disease, a chronic and systemic infection in humans and warm-blooded animals typically leading to progressive dilated cardiomyopathy and gastrointestinal manifestations. In the present study, we report that the transcription factor STAT1 (signal transducer and activator of transcription 1) reduces the susceptibility of human cells to infection with T. cruzi. Our in vitro data demonstrate that interferon -γ (IFNγ) pre-treatment causes T. cruzi-infected cells to enter an anti-parasitic state through the activation of the transcription factor STAT1. Whereas stimulation of STAT1-expressing cells with IFNγ significantly impaired intracellular replication of parasites, no protective effect of IFNγ was observed in STAT1-deficient U3A cells. The gene encoding indoleamine 2, 3-dioxygenase (ido) was identified as a STAT1-regulated target gene engaged in parasite clearance. Exposure of cells to T. cruzi trypomastigotes in the absence of IFNγ resulted in both sustained tyrosine and serine phosphorylation of STAT1 and its increased DNA binding. Furthermore, we found that in response to T. cruzi the total amount of intracellular STAT1 increased in an infectious dose-dependent manner, both at the mRNA and protein level. While STAT1 activation is a potent strategy of the host in the fight against the invading pathogen, amastigotes replicating intracellularly antagonize this pathway by specifically promoting the dephosphorylation of STAT1 serine 727, thereby partially circumventing its protective effects. These findings point to the crucial role of the IFNγ/STAT1 signal pathway in the evolutionary combat between T. cruzi parasites and their host.
Collapse
Affiliation(s)
- Philipp Stahl
- Institut für Virologie, AG Parasitologie, Philipps-Universität Marburg, Marburg, Germany
| | - Volker Ruppert
- Klinik für Kardiologie, Philipps-Universität Marburg, Marburg, Germany
| | - Ralph T. Schwarz
- Institut für Virologie, AG Parasitologie, Philipps-Universität Marburg, Marburg, Germany
- Unité de Glycobiologie Structurale et Fonctionnelle, UMR CNRS/USTL n° 8576, Université de Lille1 Sciences et Technologies, Villeneuve d'Ascq, France
| | - Thomas Meyer
- Klinik für Psychosomatische Medizin und Psychotherapie, Georg-August-Universität Göttingen, Göttingen, Germany
- * E-mail:
| |
Collapse
|
49
|
Cunha-Neto E, Chevillard C. Chagas disease cardiomyopathy: immunopathology and genetics. Mediators Inflamm 2014; 2014:683230. [PMID: 25210230 PMCID: PMC4152981 DOI: 10.1155/2014/683230] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/05/2014] [Accepted: 08/05/2014] [Indexed: 02/06/2023] Open
Abstract
Chagas disease, caused by the protozoan Trypanosoma cruzi, is endemic in Latin America and affects ca. 10 million people worldwide. About 30% of Chagas disease patients develop chronic Chagas disease cardiomyopathy (CCC), a particularly lethal inflammatory cardiomyopathy that occurs decades after the initial infection, while most patients remain asymptomatic. Mortality rate is higher than that of noninflammatory cardiomyopathy. CCC heart lesions present a Th1 T-cell-rich myocarditis, with cardiomyocyte hypertrophy and prominent fibrosis. Data suggest that the myocarditis plays a major pathogenetic role in disease progression. Major unmet goals include the thorough understanding of disease pathogenesis and therapeutic targets and identification of prognostic genetic factors. Chagas disease thus remains a neglected disease, with no vaccines or antiparasitic drugs proven efficient in chronically infected adults, when most patients are diagnosed. Both familial aggregation of CCC cases and the fact that only 30% of infected patients develop CCC suggest there might be a genetic component to disease susceptibility. Moreover, previous case-control studies have identified some genes associated to human susceptibility to CCC. In this paper, we will review the immunopathogenesis and genetics of Chagas disease, highlighting studies that shed light on the differential progression of Chagas disease patients to CCC.
Collapse
Affiliation(s)
- Edecio Cunha-Neto
- Heart Institute (InCor), University of São Paulo School of Medicine, Avenida Dr. Enéas de Carvalho Aguiar, 44 Bloco 2 9° Andar, 05406-000 São Paulo, SP, Brazil
- Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
- Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, 05406-000 São Paulo, SP, Brazil
| | | |
Collapse
|
50
|
Myocardial gene expression of T-bet, GATA-3, Ror-γt, FoxP3, and hallmark cytokines in chronic Chagas disease cardiomyopathy: an essentially unopposed TH1-type response. Mediators Inflamm 2014; 2014:914326. [PMID: 25152568 PMCID: PMC4134835 DOI: 10.1155/2014/914326] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 07/08/2014] [Indexed: 11/17/2022] Open
Abstract
Background. Chronic Chagas disease cardiomyopathy (CCC), a late consequence of Trypanosoma cruzi infection, is an inflammatory cardiomyopathy with prognosis worse than those of noninflammatory etiology (NIC). Although the T cell-rich myocarditis is known to play a pathogenetic role, the relative contribution of each of the functional T cell subsets has never been thoroughly investigated. We therefore assessed gene expression of cytokines and transcription factors involved in differentiation and effector function of each functional T cell subset (TH1/TH2/TH17/Treg) in CCC, NIC, and heart donor myocardial samples. Methods and Results. Quantitative PCR showed markedly upregulated expression of IFN-γ and transcription factor T-bet, and minor increases of GATA-3; FoxP3 and CTLA-4; IL-17 and IL-18 in CCC as compared with NIC samples. Conversely, cytokines expressed by TH2 cells (IL-4, IL-5, and IL-13) or associated with Treg (TGF-β
and IL-10) were not upregulated in CCC myocardium. Expression of TH1-related genes such as T-bet, IFN-γ, and IL-18 correlated with ventricular dilation, FoxP3, and CTLA-4. Conclusions. Results are consistent with a strong local TH1-mediated response in most samples, possibly associated with pathological myocardial remodeling, and a proportionally smaller FoxP3+CTLA4+ Treg cell population, which is unable to completely curb IFN-γ production in CCC myocardium, therefore fueling inflammation.
Collapse
|