1
|
Pacini MF, Bulfoni Balbi C, Dinatale B, Farré C, Cacik P, Gonzalez FB, Marcipar I, Pérez AR. Intranasal Trans-Sialidase Vaccine Mitigates Acute and Chronic Pathology in a Preclinical Oral Chagas Disease Model. Vaccines (Basel) 2024; 12:1171. [PMID: 39460337 PMCID: PMC11511307 DOI: 10.3390/vaccines12101171] [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: 09/13/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Chagas disease, caused by Trypanosoma cruzi, leads to severe complications in 30% of infected individuals, including acute myocarditis and chronic fibrosing cardiomyopathy. Despite the significant burden of this disease, there is currently no licensed vaccine available to prevent it. This study aimed to evaluate the mucosal and systemic immunogenicity as well as the prophylactic efficacy of a mucosal vaccine candidate and its impact on both acute and chronic cardiomyopathy. The results showed that the nasal administration of trans-sialidase (TS) plus c-di-AMP (TS+A) vaccine elicited a NALT expression of IFN-γ, IL-17a and IL-4 mRNA as well as a nasal-specific production of IgA. An in vivo challenge with TS also triggered increased proliferation of lymphocytes from the NALT, sentinel cervical lymph node, and spleen. TS+A immunization increased the plasma levels of Th1/Th2/Th17 cytokines and elicited an evident cellular response by which to judge enhanced delayed-type hypersensitivity responses following a TS footpad challenge. After oral infection, TS+A-vaccinated mice showed significantly reduced parasitemia and parasite load in the heart, muscles and intestines, while markers of hepatic and muscle damage as well as clinical manifestations of acute infection were strongly diminished. TS+A also attenuated acute myocarditis and the expression of inflammatory markers in the heart. The protection conferred by TS+A extended into the chronic phase, where it resulted in a clear reduction in chronic myocarditis, fibrosis and functional electrocardiographic abnormalities, associated with a decreased expression of the pro-fibrotic TGF-β. These results revealed that it is possible to develop a mucosal vaccine against T. cruzi based on TS and c-di-AMP that is capable of reducing the development of Chagas cardiomyopathy, the hallmark of Chagas disease.
Collapse
Affiliation(s)
- Maria Florencia Pacini
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario 2000, Argentina; (M.F.P.); (C.B.B.); (B.D.); (C.F.); (F.B.G.)
| | - Camila Bulfoni Balbi
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario 2000, Argentina; (M.F.P.); (C.B.B.); (B.D.); (C.F.); (F.B.G.)
| | - Brenda Dinatale
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario 2000, Argentina; (M.F.P.); (C.B.B.); (B.D.); (C.F.); (F.B.G.)
| | - Cecilia Farré
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario 2000, Argentina; (M.F.P.); (C.B.B.); (B.D.); (C.F.); (F.B.G.)
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario 2000, Argentina
| | - Paula Cacik
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe 3000, Argentina; (P.C.); (I.M.)
| | - Florencia Belén Gonzalez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario 2000, Argentina; (M.F.P.); (C.B.B.); (B.D.); (C.F.); (F.B.G.)
| | - Iván Marcipar
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe 3000, Argentina; (P.C.); (I.M.)
| | - Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER-CONICET), Rosario 2000, Argentina; (M.F.P.); (C.B.B.); (B.D.); (C.F.); (F.B.G.)
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario 2000, Argentina
| |
Collapse
|
2
|
Lozano KJG, Gonçalves Santos E, Vilas Boas DF, Oliveira RRG, Diniz LF, Benedetti MD, Carneiro CM, C Bandeira L, Faria G, Gonçalves RV, Novaes RD, Caldas S, Caldas IS. Schistosoma mansoni co-infection modulates Chagas disease development but does not impair the effect of benznidazole-based chemotherapy. Int Immunopharmacol 2024; 128:111467. [PMID: 38211479 DOI: 10.1016/j.intimp.2023.111467] [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: 08/24/2023] [Revised: 12/05/2023] [Accepted: 12/27/2023] [Indexed: 01/13/2024]
Abstract
The adequate management of parasite co-infections represents a challenge that has not yet been overcome, especially considering that the pathological outcomes and responses to treatment are poorly understood. Thus, this study aimed to evaluate the impact of Schistosoma mansoni infection on the efficacy of benznidazole (BZN)-based chemotherapy in Trypanosoma cruzi co-infected mice. BALB/c mice were maintained uninfected or co-infected with S. mansoni and T. cruzi, and were untreated or treated with BZN. Body weight, mortality, parasitemia, cardiac parasitism, circulating cytokines (Th1/Th2/Th17); as well as heart, liver and intestine microstructure were analyzed. The parasitemia peak was five times higher and myocarditis was more severe in co-infected than T. cruzi-infected mice. After reaching peak, parasitemia was effectively controlled in co-infected animals. BZN successfully controlled parasitemia in both co-infected and T. cruzi-infected mice and improved body mass, cardiac parasitism, myocarditis and survival in co-infected mice. Co-infection dampened the typical cytokine response to either parasite, and BZN reduced anti-inflammatory cytokines in co-infected mice. Despite BZN normalizing splenomegaly and liver cellular infiltration, it exacerbated hepatomegaly in co-infected mice. Co-infection or BZN exerted no effect on hepatic granulomas, but increased pulmonary and intestinal granulomas. Marked granulomatous inflammation was identified in the small intestine of all schistosomiasis groups. Taken together, our findings indicate that BZN retains its therapeutic efficacy against T. cruzi infection even in the presence of S. mansoni co-infection, but with organ-specific repercussions, especially in the liver.
Collapse
Affiliation(s)
- Kelly J G Lozano
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Elda Gonçalves Santos
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Diego F Vilas Boas
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Raphaela R G Oliveira
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Lívia F Diniz
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Monique D Benedetti
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Cláudia M Carneiro
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University, Ouro Preto 35400-000, MG, Brazil
| | - Lorena C Bandeira
- Laboratory of Immunopathology, Nucleus of Biological Sciences Research, Federal University, Ouro Preto 35400-000, MG, Brazil
| | - Gilson Faria
- Department of Research and Development., Ezequiel Dias Foundation, 30510-010, Belo Horizonte, MG, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa 36570-900, MG, Brazil
| | - Rômulo D Novaes
- Department of Structural Biology, Federal University of Alfenas, Alfenas 37130-000, MG, Brazil
| | - Sérgio Caldas
- Department of Research and Development., Ezequiel Dias Foundation, 30510-010, Belo Horizonte, MG, Brazil
| | - Ivo S Caldas
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil.
| |
Collapse
|
3
|
Reis RCFM, Dos Santos EG, Benedetti MD, Reis ACC, Brandão GC, Silva GND, Diniz LA, Ferreira RS, Caldas IS, Braga SFP, Souza TBD. Design and synthesis of new 1,2,3-triazoles derived from eugenol and analogues with in vitro and in vivo activity against Trypanosoma cruzi. Eur J Med Chem 2023; 258:115622. [PMID: 37441850 DOI: 10.1016/j.ejmech.2023.115622] [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: 05/17/2023] [Revised: 06/22/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023]
Abstract
Chagas disease (CD) is a neglected tropical disease endemic in 21 countries and affects about 8 million people around the world. The pharmacotherapy for this disease is limited to two drugs (Benznidazole and Nifurtimox) and both are associated with important limitations, as low cure rate in the chronic phase of the disease, high toxicity and increasing resistance by Trypanosoma cruzi. Recently, we reported a bioactive 1,2,3-triazole (compound 35) active in vitro (IC50 42.8 μM) and in vivo (100 mg/kg) against T. cruzi Y strains and preliminary in silico studies suggested the cysteine protease cruzain as a possible target. Considering these initial findings, we describe here the design and synthesis of new 1,2,3-triazoles derivatives of our hit compound (35). The triazoles were initially evaluated against healthy cells derived from neonatal rat cardiomyoblasts (H9c2 cells) to determine their cytotoxicity and against epimastigotes forms of T. cruzi Y strain. The most active triazoles were compounds 26 (IC50 19.7 μM) and 27 (IC50 7.3 μM), while benznidazole was active at 21.6 μM. Derivative 27 showed an interesting selectivity index considering healthy H9c2 cells (>77). Promising activities against trypomastigotes forms of the parasite were also observed for triazoles 26 (IC50 20.74 μM) and 27 (IC50 8.41 μM), mainly 27 which showed activity once again higher than that observed for benznidazole (IC50 12.72 μM). While docking results suggested cruzain as a potential target for these compounds, no significant enzyme inhibition was observed in vitro, indicating that their trypanocidal activity is related to another mode of action. Considering the promising in vitro results of triazoles 26 and 27, the in vivo toxicity was initially verified based on the evaluation of behavioral and physiological parameters, mortality, effect in body weight gain, and through the measurement of AST/ALT enzymes, which are markers of liver toxicity. All these evaluations pointed to a good tolerability of the animals, especially considering triazole 27. A reduction in parasitemia was observed among animals treated with triazole 27, but not among those treated with derivative 26. Regarding the dosage, derivative 27 (100 mg/kg) was the most active sample against T. cruzi infection, showing a 99.4% reduction in parasitemia peak. Triazole 27 at a dosage of 100 mg/kg influenced the humoral immune response and reduced myocarditis in the animals, bringing antibody levels closer to those observed among healthy mice. Altogether, our results indicate compound 27 as a new lead for the development of drug candidates to treat Chagas disease.
Collapse
Affiliation(s)
| | - Elda Gonçalves Dos Santos
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | - Monique Dias Benedetti
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | | | - Geraldo Célio Brandão
- School of Pharmacy - Federal University of Ouro Preto, 35400-000, Ouro Preto, MG, Brazil
| | | | - Lucas Abreu Diniz
- Biochemistry and Immunology Department - Federal University of Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Rafaela Salgado Ferreira
- Biochemistry and Immunology Department - Federal University of Minas Gerais, 31270-901, Belo Horizonte, MG, Brazil
| | - Ivo Santana Caldas
- Department of Pathology and Parasitology, Federal University of Alfenas, 37130-001, Alfenas, MG, Brazil
| | | | | |
Collapse
|
4
|
Kloc M, Kubiak JZ, Zdanowski R, Ghobrial RM. Memory Macrophages. Int J Mol Sci 2022; 24:ijms24010038. [PMID: 36613481 PMCID: PMC9819859 DOI: 10.3390/ijms24010038] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/12/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Immunological memory is a crucial part of the immune defense that allows organisms to respond against previously encountered pathogens or other harmful factors. Immunological memory is based on the establishment of epigenetic modifications of the genome. The ability to memorize encounters with pathogens and other harmful factors and mount enhanced defense upon subsequent encounters is an evolutionarily ancient mechanism operating in all animals and plants. However, the term immunological memory is usually restricted to the organisms (invertebrates and vertebrates) possessing the immune system. The mammalian immune system, with innate and adaptive branches, is the most sophisticated among vertebrates. The concept of innate memory and memory macrophages is relatively new and thus understudied. We introduce the concept of immunological memory and describe types of memory in different species and their evolutionary status. We discuss why the traditional view of innate immune cells as the first-line defenders is too restrictive and how the innate immune cells can accumulate and retain immunologic memory. We describe how the initial priming leads to chromatin remodeling and epigenetic changes, which allow memory macrophage formation. We also summarize what is currently known about the mechanisms underlying development of memory macrophages; their molecular and metabolic signature and surface markers; and how they may contribute to immune defense, diseases, and organ transplantation.
Collapse
Affiliation(s)
- Malgorzata Kloc
- The Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
- Correspondence:
| | - Jacek Z. Kubiak
- Dynamics and Mechanics of Epithelia Group, Faculty of Medicine, Institute of Genetics and Development of Rennes, University of Rennes, CNRS, UMR 6290, 35043 Rennes, France
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, Szaserow 128, 04-141 Warsaw, Poland
| | - Robert Zdanowski
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine National Research Institute, Szaserow 128, 04-141 Warsaw, Poland
| | - Rafik M. Ghobrial
- The Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
5
|
Savino W, Durães J, Maldonado-Galdeano C, Perdigon G, Mendes-da-Cruz DA, Cuervo P. Thymus, undernutrition, and infection: Approaching cellular and molecular interactions. Front Nutr 2022; 9:948488. [PMID: 36225882 PMCID: PMC9549110 DOI: 10.3389/fnut.2022.948488] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Undernutrition remains a major issue in global health. Low protein-energy consumption, results in stunting, wasting and/or underweight, three deleterious forms of malnutrition that affect roughly 200 million children under the age of five years. Undernutrition compromises the immune system with the generation of various degrees of immunodeficiency, which in turn, renders undernourished individuals more sensitive to acute infections. The severity of various infectious diseases including visceral leishmaniasis (VL), influenza, and tuberculosis is associated with undernutrition. Immunosuppression resulting from protein-energy undernutrition severely impacts primary and secondary lymphoid organs involved in the response to related pathogens. The thymus-a primary lymphoid organ responsible for the generation of T lymphocytes-is particularly compromised by both undernutrition and infectious diseases. In this respect, we will discuss herein various intrathymic cellular and molecular interactions seen in undernutrition alone or in combination with acute infections. Many examples illustrated in studies on humans and experimental animals clearly revealed that protein-related undernutrition causes thymic atrophy, with cortical thymocyte depletion. Moreover, the non-lymphoid microenvironmental compartment of the organ undergoes important changes in thymic epithelial cells, including their secretory products such as hormones and extracellular matrix proteins. Of note, deficiencies in vitamins and trace elements also induce thymic atrophy. Interestingly, among the molecular interactions involved in the control of undernutrition-induced thymic atrophy is a hormonal imbalance with a rise in glucocorticoids and a decrease in leptin serum levels. Undernutrition also yields a negative impact of acute infections upon the thymus, frequently with the intrathymic detection of pathogens or their antigens. For instance, undernourished mice infected with Leishmania infantum (that causes VL) undergo drastic thymic atrophy, with significant reduction in thymocyte numbers, and decreased levels of intrathymic chemokines and cytokines, indicating that both lymphoid and microenvironmental compartments of the organ are affected. Lastly, recent data revealed that some probiotic bacteria or probiotic fermented milks improve the thymus status in a model of malnutrition, thus raising a new field for investigation, namely the thymus-gut connection, indicating that probiotics can be envisioned as a further adjuvant therapy in the control of thymic changes in undernutrition accompanied or not by infection.
Collapse
Affiliation(s)
- Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Jonathan Durães
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory on Leishmaniasis Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Carolina Maldonado-Galdeano
- Laboratory of Immunology, Reference Center for Lactobacilli Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucumán, Argentina
- Laboratory of Immunology, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucumán, San Miguel de Tucumán, Argentina
| | - Gabriela Perdigon
- Laboratory of Immunology, Reference Center for Lactobacilli Centro de Referencia para Lactobacilos-Consejo Nacional de Investigaciones Científicas y Técnicas (CERELA-CONICET), San Miguel de Tucumán, Argentina
- Laboratory of Immunology, Faculty of Biochemistry, Chemistry and Pharmacy, National University of Tucumán, San Miguel de Tucumán, Argentina
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, United Kingdom
| | - Patricia Cuervo
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory on Leishmaniasis Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Gómez I, Thomas MC, Palacios G, Egui A, Carrilero B, Simón M, Valladares B, Segovia M, Carmelo E, López MC. Differential Expression of Immune Response Genes in Asymptomatic Chronic Chagas Disease Patients Versus Healthy Subjects. Front Cell Infect Microbiol 2021; 11:722984. [PMID: 34552885 PMCID: PMC8450343 DOI: 10.3389/fcimb.2021.722984] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/12/2021] [Indexed: 11/30/2022] Open
Abstract
Infection by the Trypanosoma cruzi parasite causes Chagas disease and triggers multiple immune mechanisms in the host to combat the pathogen. Chagas disease has a variable clinical presentation and progression, producing in the chronic phase a fragile balance between the host immune response and parasite replication that keeps patients in a clinically silent asymptomatic stage for years. Since the parasite is intracellular and replicates within cells, the cell-mediated response of the host adaptive immunity plays a critical role. This function is mainly orchestrated by T lymphocytes, which recognize parasite antigens and promote specific functions to control the infection. However, little is known about the immunological markers associated with this asymptomatic stage of the disease. In this large-scale analysis, the differential expression of 106 immune system-related genes has been analyzed using high-throughput qPCR in T. cruzi antigen-stimulated PBMC from chronic Chagas disease patients with indeterminate form (IND) and healthy donors (HD) from endemic and non-endemic areas of Chagas disease. This analysis revealed that there were no differences in the expression level of most genes under study between healthy donors from endemic and non-endemic areas determined by PCA and differential gene expression analysis. Instead, PCA revealed the existence of different expression profiles between IND patients and HD (p < 0.0001), dependent on the 32 genes included in PC1. Differential gene expression analysis also revealed 23 upregulated genes (expression fold change > 2) and 11 downregulated genes (expression fold change < 0.5) in IND patients versus HD. Enrichment analysis showed that several upregulated genes in IND patients participate in relevant immunological pathways such as antigen-dependent B cell activation, stress induction of HSP regulation, NO2-dependent IL12 pathway in NK cells, and cytokine-inflammatory response. The antigen-specific differential gene expression profile detected in these patients and the relevant immunological pathways that seem to be activated could represent potential biomarkers of the asymptomatic form of Chagas disease, helpful to diagnosis and infection control.
Collapse
Affiliation(s)
- Inmaculada Gómez
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - M Carmen Thomas
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Génesis Palacios
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| | - Adriana Egui
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Bartolomé Carrilero
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Marina Simón
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Basilio Valladares
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain.,Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, La Laguna, Spain
| | - Manuel Segovia
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Emma Carmelo
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain.,Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, La Laguna, Spain
| | - Manuel Carlos López
- Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
7
|
Luo M, Xu L, Qian Z, Sun X. Infection-Associated Thymic Atrophy. Front Immunol 2021; 12:652538. [PMID: 34113341 PMCID: PMC8186317 DOI: 10.3389/fimmu.2021.652538] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
The thymus is a vital organ of the immune system that plays an essential role in thymocyte development and maturation. Thymic atrophy occurs with age (physiological thymic atrophy) or as a result of viral, bacterial, parasitic or fungal infection (pathological thymic atrophy). Thymic atrophy directly results in loss of thymocytes and/or destruction of the thymic architecture, and indirectly leads to a decrease in naïve T cells and limited T cell receptor diversity. Thus, it is important to recognize the causes and mechanisms that induce thymic atrophy. In this review, we highlight current progress in infection-associated pathogenic thymic atrophy and discuss its possible mechanisms. In addition, we discuss whether extracellular vesicles/exosomes could be potential carriers of pathogenic substances to the thymus, and potential drugs for the treatment of thymic atrophy. Having acknowledged that most current research is limited to serological aspects, we look forward to the possibility of extending future work regarding the impact of neural modulation on thymic atrophy.
Collapse
Affiliation(s)
- Mingli Luo
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lingxin Xu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zhengyu Qian
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Xi Sun
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| |
Collapse
|
8
|
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
|
9
|
CD8low T cells expanded following acute Trypanosoma cruzi infection and benznidazole treatment are a relevant subset of IFN-γ producers. PLoS Negl Trop Dis 2020; 14:e0008969. [PMID: 33347472 PMCID: PMC7785226 DOI: 10.1371/journal.pntd.0008969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/05/2021] [Accepted: 11/10/2020] [Indexed: 11/30/2022] Open
Abstract
CD8 T cells are regarded as pivotal players in both immunoprotection and immunopathology following Trypanosoma cruzi infection. Previously, we demonstrated the expansion of CD8+ T lymphocytes in the spleen of T. cruzi-infected mice under treatment with benznidazole (N-benzyl-2-nitroimidazole acetamide; Bz), a drug available for clinical therapy. This finding underlies the concept that the beneficial effects of Bz on controlling acute T. cruzi infection are related to a synergistic process between intrinsic trypanocidal effect and indirect triggering of the active immune response. In the present study, we particularly investigated the effect of Bz treatment on the CD8+ T cell subset following T. cruzi infection. Herein we demonstrated that, during acute T. cruzi infection, Bz treatment reduces and abbreviates the parasitemia, but maintains elevated expansion of CD8+ T cells. Within this subset, a remarkable group of CD8low cells was found in both Bz-treated and non-treated infected mice. In Bz-treated mice, early pathogen control paralleled the lower frequency of recently activated CD8low cells, as ascertained by CD69 expression. However, the CD8low subset sustains significant levels of CD44highCD62Llow and CD62LlowT-bethigh effector memory T cells, in both Bz-treated and non-treated infected mice. These CD8low cells also comprise the main group of spontaneous interferon (IFN)-γ-producing CD8+ T cells. Interestingly, following in vitro anti-CD3/CD28 stimulation, CD8+ T cells from Bz-treated T. cruzi-infected mice exhibited higher frequency of IFN-γ+ cells, which bear mostly a CD8low phenotype. Altogether, our results point to the marked presence of CD8low T cells that arise during acute T. cruzi infection, with Bz treatment promoting their significant expansion along with a potential effector program for high IFN-γ production. Chagas disease is a neglected illness caused by the protozoan Trypanosoma cruzi, which affects 6 to 7 million people worldwide. The current treatment for acutely-infected patients is mostly limited to the benznidazole (Bz) drug, reaching up to 80% of cure. It has been proposed that Bz therapy efficacy involves both trypanocidal and immunonodulatory effects. In this sense, we previously suggested that CD8+ T cells, highly expanded after Bz treatment of acute T. cruzi-infected mice, might play a particular role in parasite control. Here, by further investigating those expanded CD8+ T cells, we observed that they bear a clear-cut effector phenotype and that a significant part of them stand out as a subpopulation bearing low levels of CD8 on their surface. Interestingly, besides the evident parasite control, Bz-treated mice sustain a group of effector CD8low cells with spontaneous IFN-γ production. Moreover, in vitro-stimulated CD8+ T cells, sorted from infected and Bz-treated mice, present a relevant group of IFN-γ+ cells with a CD8low phenotype. Altogether, our data indicate the particular subset of CD8low cells as potentially contributing for a sustained protective immunity in T. cruzi-infected animals under Bz therapy.
Collapse
|
10
|
Hypothyroidism impairs the host immune response during the acute phase of Chagas disease. Immunobiology 2020; 225:152024. [PMID: 33227693 DOI: 10.1016/j.imbio.2020.152024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/20/2020] [Accepted: 10/18/2020] [Indexed: 11/24/2022]
Abstract
Diseases associated with thyroid hypofunction have been the subject of studies in infectious models, since several authors have demonstrated a pivotal role of iodinated hormones (thyroxine and triiodothyronine) in the modulation of immune effector responses. Using a model of hypothyroidism induced by anti-thyroid drug, we investigated the influence of hypothyroidism in the course of acute Trypanosoma cruzi infection. For this, male Hannover Wistar rats were challenged with methimazole for 21 days (0.02% in drinking water), and water for control counterparts. After confirmation of the hypothyroidism, rats were intraperitoneally challenged with 1x105 blood trypomastigotes of the Y strain of T. cruzi. Our findings suggest that hypothyroidism impairs animal weight gain, but does not affect the health of essential organs. Interestingly, infected hypothyroid animals had a significant increase in thymic cell death, with consequent drop in lymphocyte frequency in whole blood (evaluated on the 11th day of infection). Analyzing the percentage of immune cells in the spleen, we found a strong influence of hypothyroidism as a negative regulator of B cells, and antigenic ability of macrophages (RT1b expression) in the course of the experimental chagasic infection. Enhanced serum IL-17A concentration was induced by T. cruzi infection, but hypothyroidism impaired the production of this mediator as seen in infected hypothyroid animals. Taken together, our work suggests for the first time that hypothyroidism may adversely interfere with the modulation of effective immunity in the early phase of Chagas' disease.
Collapse
|
11
|
Pérez AR, de Meis J, Rodriguez-Galan MC, Savino W. The Thymus in Chagas Disease: Molecular Interactions Involved in Abnormal T-Cell Migration and Differentiation. Front Immunol 2020; 11:1838. [PMID: 32983098 PMCID: PMC7492291 DOI: 10.3389/fimmu.2020.01838] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022] Open
Abstract
Chagas disease, caused by the protozoan parasite T. cruzi, is a prevalent parasitic disease in Latin America. Presently, it is spreading around the world by human migration, thus representing a new global health issue. Chronically infected individuals reveal a dissimilar disease progression: while nearly 60% remain without apparent disease for life, 30% develop life-threatening pathologies, such as chronic chagasic cardiomyopathy (CCC) or megaviscerae. Inflammation driven by parasite persistence seems to be involved in the pathophysiology of the disease. However, there is also evidence of the occurrence of autoimmune events, mainly caused by molecular mimicry and bystander activation. In experimental models of disease, is well-established that T. cruzi infects the thymus and causes locally profound structural and functional alterations. The hallmark is a massive loss of CD4+CD8+ double positive (DP) thymocytes, mainly triggered by increased levels of glucocorticoids, although other mechanisms seem to act simultaneously. Thymic epithelial cells (TEC) exhibited an increase in extracellular matrix deposition, which are related to thymocyte migratory alterations. Moreover, medullary TEC showed a decreased expression of AIRE and altered expression of microRNAs, which might be linked to a disrupted negative selection of the T-cell repertoire. Also, almost all stages of thymocyte development are altered, including an abnormal output of CD4−CD8− double negative (DN) and DP immature and mature cells, many of them carrying prohibited TCR-Vβ segments. Evidence has shown that DN and DP cells with an activated phenotype can be tracked in the blood of humans with chronic Chagas disease and also in the secondary lymphoid organs and heart of infected mice, raising new questions about the relevance of these populations in the pathogenesis of Chagas disease and their possible link with thymic alterations and an immunoendocrine imbalance. Here, we discuss diverse molecular mechanisms underlying thymic abnormalities occurring during T. cruzi infection and their link with CCC, which may contribute to the design of innovative strategies to control Chagas disease pathology.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario, CONICET-Universidad Nacional de Rosario, Rosario, Argentina.,Centro de Investigación y Producción de Reactivos Biológicos, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Ferreira RR, Abreu RDS, Vilar-Pereira G, Degrave W, Meuser-Batista M, Ferreira NVC, da Cruz Moreira O, da Silva Gomes NL, Mello de Souza E, Ramos IP, Bailly S, Feige JJ, Lannes-Vieira J, de Araújo-Jorge TC, Waghabi MC. TGF-β inhibitor therapy decreases fibrosis and stimulates cardiac improvement in a pre-clinical study of chronic Chagas' heart disease. PLoS Negl Trop Dis 2019; 13:e0007602. [PMID: 31365537 PMCID: PMC6690554 DOI: 10.1371/journal.pntd.0007602] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 08/12/2019] [Accepted: 07/03/2019] [Indexed: 01/11/2023] Open
Abstract
TGF-β involvement in Chagas disease cardiomyopathy has been clearly demonstrated. The TGF-β signaling pathway is activated in the cardiac tissue of chronic phase patients and is associated with an increase in extracellular matrix protein expression. The aim of this study was to investigate the effect of GW788388, a selective inhibitor of TβR1/ALK5, on cardiac function in an experimental model of chronic Chagas' heart disease. To this end, C57BL/6 mice were infected with Trypanosoma cruzi (102 parasites from the Colombian strain) and treated orally with 3mg/kg GW788388 starting at 120 days post-infection (dpi), when 100% of the infected mice show cardiac damage, and following three distinct treatment schedules: i) single dose; ii) one dose per week; or iii) three doses per week during 30 days. The treatment with GW788388 improved several cardiac parameters: reduced the prolonged PR and QTc intervals, increased heart rate, and reversed sinus arrhythmia, and atrial and atrioventricular conduction disorders. At 180 dpi, 30 days after treatment interruption, the GW3x-treated group remained in a better cardiac functional condition. Further, GW788388 treatment reversed the loss of connexin-43 enriched intercellular plaques and reduced fibrosis of the cardiac tissue. Inhibition of the TGF-β signaling pathway reduced TGF-β/pSmad2/3, increased MMP-9 and Sca-1, reduced TIMP-1/TIMP-2/TIMP-4, and partially restored GATA-6 and Tbox-5 transcription, supporting cardiac recovery. Moreover, GW788388 administration did not modify cardiac parasite load during the infection but reduced the migration of CD3+ cells to the heart tissue. Altogether, our data suggested that the single dose schedule was not as effective as the others and treatment three times per week during 30 days seems to be the most effective strategy. The therapeutic effects of GW788388 are promising and suggest a new possibility to treat cardiac fibrosis in the chronic phase of Chagas' heart disease by TGF-β inhibitors.
Collapse
Affiliation(s)
- Roberto Rodrigues Ferreira
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Rayane da Silva Abreu
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Glaucia Vilar-Pereira
- Laboratório de Biologia das Interações—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Wim Degrave
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Marcelo Meuser-Batista
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
- Departamento de Anatomia Patológica e Citopatologia, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brasil
| | - Nilma Valéria Caldeira Ferreira
- Departamento de Anatomia Patológica e Citopatologia, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brasil
| | - Otacílio da Cruz Moreira
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz (FIOCRUZ/RJ), Rio de Janeiro, Brazil
| | - Natália Lins da Silva Gomes
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz (FIOCRUZ/RJ), Rio de Janeiro, Brazil
| | - Elen Mello de Souza
- Laboratório de Virologia Molecular—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Isalira P. Ramos
- UFRJ, Centro Nacional de Biologia Estrutural e Bioimagem, Rio de Janeiro, RJ, Brazil
| | - Sabine Bailly
- Université Grenoble-Alpes, Inserm, CEA, Biology of Cancer and Infection Laboratory, Grenoble, France
| | - Jean-Jacques Feige
- Université Grenoble-Alpes, Inserm, CEA, Biology of Cancer and Infection Laboratory, Grenoble, France
| | - Joseli Lannes-Vieira
- Laboratório de Biologia das Interações—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Tania C. de Araújo-Jorge
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
| | - Mariana Caldas Waghabi
- Laboratório de Genômica Funcional e Bioinformática—Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro RJ, Brasil
- * E-mail:
| |
Collapse
|
13
|
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: 94] [Impact Index Per Article: 18.8] [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
|
14
|
Baez NS, Cerbán F, Savid-Frontera C, Hodge DL, Tosello J, Acosta-Rodriguez E, Almada L, Gruppi A, Viano ME, Young HA, Rodriguez-Galan MC. Thymic expression of IL-4 and IL-15 after systemic inflammatory or infectious Th1 disease processes induce the acquisition of "innate" characteristics during CD8+ T cell development. PLoS Pathog 2019; 15:e1007456. [PMID: 30608984 PMCID: PMC6319713 DOI: 10.1371/journal.ppat.1007456] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 11/05/2018] [Indexed: 01/28/2023] Open
Abstract
Innate CD8+ T cells express a memory-like phenotype and demonstrate a strong cytotoxic capacity that is critical during the early phase of the host response to certain bacterial and viral infections. These cells arise in the thymus and depend on IL-4 and IL-15 for their development. Even though innate CD8+ T cells exist in the thymus of WT mice in low numbers, they are highly enriched in KO mice that lack certain kinases, leading to an increase in IL-4 production by thymic NKT cells. Our work describes that in C57BL/6 WT mice undergoing a Th1 biased infectious disease, the thymus experiences an enrichment of single positive CD8 (SP8) thymocytes that share all the established phenotypical and functional characteristics of innate CD8+ T cells. Moreover, through in vivo experiments, we demonstrate a significant increase in survival and a lower parasitemia in mice adoptively transferred with SP8 thymocytes from OT I—T. cruzi-infected mice, demonstrating that innate CD8+ thymocytes are able to protect against a lethal T. cruzi infection in an Ag-independent manner. Interestingly, we obtained similar results when using thymocytes from systemic IL-12 + IL-18-treated mice. This data indicates that cytokines triggered during the acute stage of a Th1 infectious process induce thymic production of IL-4 along with IL-15 expression resulting in an adequate niche for development of innate CD8+ T cells as early as the double positive (DP) stage. Our data demonstrate that the thymus can sense systemic inflammatory situations and alter its conventional CD8 developmental pathway when a rapid innate immune response is required to control different types of pathogens. Murine innate CD8+ T cells demonstrate strong cytotoxic capacity during the early phase of certain bacterial and viral infections. Such cells have been reported to be present in both mice and humans but many questions remain as to their differentiation and maturation process. Innate CD8+ T cells arise in the thymus and depend on IL-4 and IL-15 for their development. A description of the cellular and molecular mechanisms involved during their thymic development has been obtained from KO mice that lack kinases and transcription factors important for TCR signaling. In these mice, SP8 thymocytes with an innate phenotype are highly enriched over the conventional SP8 cells. Our work describes, for the first time, that in WT mice, thymic IL-4 and IL-15 expression triggered by Th1 infectious processes induce an adequate niche for development of innate rather than conventional CD8+ T cells. Our data show that the thymus is able to sense a systemic inflammatory response (probably mediated by systemic IL-12 and IL-18 production) and alter its ontogeny when pathogen control is needed.
Collapse
Affiliation(s)
- Natalia S. Baez
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fabio Cerbán
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Constanza Savid-Frontera
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Deborah L. Hodge
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Jimena Tosello
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Eva Acosta-Rodriguez
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Laura Almada
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Adriana Gruppi
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Estefania Viano
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Howard A. Young
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Maria Cecilia Rodriguez-Galan
- Inmunología. CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
15
|
Barreto de Albuquerque J, Silva Dos Santos D, Stein JV, de Meis J. Oral Versus Intragastric Inoculation: Similar Pathways of Trypanosoma cruzi Experimental Infection? From Target Tissues, Parasite Evasion, and Immune Response. Front Immunol 2018; 9:1734. [PMID: 30100907 PMCID: PMC6072848 DOI: 10.3389/fimmu.2018.01734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
Currently, oral infection is the most frequent transmission mechanism of Chagas disease in Brazil and others Latin American countries. This transmission pathway presents increased mortality rate in the first 2 weeks, which is higher than the calculated mortality after the biting of infected insect vectors. Thus, the oral route of Trypanosoma cruzi infection, and the consequences in the host must be taken into account when thinking on the mechanisms underlying the natural history of the disease. Distinct routes of parasite entry may differentially affect immune circuits, stimulating regional immune responses that impact on the overall profile of the host protective immunity. Experimental studies related to oral infection usually comprise inoculation in the mouth (oral infection, OI) or gavage (gastrointestinal infection, GI), being often considered as similar routes of infection. Hence, establishing a relationship between the inoculation site (OI or GI) with disease progression and the mounting of T. cruzi-specific regional immune responses is an important issue to be considered. Here, we provide a discussion on studies performed in OI and GI in experimental models of acute infections, including T. cruzi infection.
Collapse
Affiliation(s)
| | - Danielle Silva Dos Santos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Perez CJ, Thompson RCA, Keatley SK, Walsh AL, Lymbery AJ. The effect of reinfection and mixed Trypanosoma cruzi infections on disease progression in mice. Acta Trop 2018; 178:107-114. [PMID: 29113781 DOI: 10.1016/j.actatropica.2017.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 10/17/2017] [Accepted: 11/03/2017] [Indexed: 12/23/2022]
Abstract
The progression of Chagas disease (CD) varies significantly from host to host and is affected by multiple factors. In particular, mixed strain infections and reinfections have the potential to exacerbate disease progression subsequently affecting clinical management of patients with CD. Consequently, an associated reduction in therapeutic intervention and poor prognosis may occur due to this exacerbated disease state. This study investigated the effects of mixed strain infections and reinfection with Trypanosoma cruzi in mice, using two isolates from different discrete typing units, TcI (C8 clone 1) and TcIV (10R26). There were no significant differences in mortality rate, body weight or body condition among mice infected with either C8 clone 1, 10R26, or a mixture of both isolates. However, the parasite was found in a significantly greater number of host organs in mice infected with a mixture of isolates, and the histopathological response to infection was significantly greater in mice infected with C8 clone 1 alone, and C8 clone 1+10R26 mixed infections than in mice infected with 10R26 alone. To investigate the effects of reinfection, mice received either a double exposure to C8 clone 1; a double exposure to 10R26; exposure to C8 clone 1 followed by 10R26; or exposure to 10R26 followed by C8 clone 1. Compared to single infection groups, mortality was significantly increased, while survival time, body weight and body condition were all significantly decreased across all reinfection groups, with no significant differences among these groups. The mortality rate over all reinfection groups was 63.6%, compared to 0% in single infection groups, however there was no evidence of a greater histopathological response to infection. These results suggest firstly, that the C8 clone 1 isolate is more virulent than the 10R26 isolate, and secondly, that a more disseminated infection may occur with a mixture of isolates than with single isolates, although there is no evidence that mixed infections have a greater pathological effect. By contrast, reinfections do have major effects on host survivability and thus disease outcome. This confirms previous research demonstrating spontaneous deaths following reinfection, a phenomenon that to our knowledge has only been reported once before.
Collapse
Affiliation(s)
- Catherine J Perez
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch 6150, Australia.
| | - R C Andrew Thompson
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch 6150, Australia.
| | - Sarah K Keatley
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch 6150, Australia.
| | - Audra L Walsh
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch 6150, Australia.
| | - Alan J Lymbery
- School of Veterinary and Life Sciences, Murdoch University, South Street, Murdoch 6150, Australia; Freshwater Fish Group and Fish Health Unit, Murdoch University,South Street, Murdoch 6150, Australia.
| |
Collapse
|
17
|
Pérez AR, Morrot A, Carvalho VF, de Meis J, Savino W. Role of Hormonal Circuitry Upon T Cell Development in Chagas Disease: Possible Implications on T Cell Dysfunctions. Front Endocrinol (Lausanne) 2018; 9:334. [PMID: 29963015 PMCID: PMC6010535 DOI: 10.3389/fendo.2018.00334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/31/2018] [Indexed: 12/22/2022] Open
Abstract
T cell response plays an essential role in the host resistance to infection by the protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease. This infection is often associated with multiple manifestations of T cell dysfunction, both during the acute and the chronic phases of disease. Additionally, the normal development of T cells is affected. As seen in animal models of Chagas disease, there is a strong thymic atrophy due to massive death of CD4+CD8+ double-positive cells by apoptosis and an abnormal escape of immature and potentially autoreactive thymocytes from the organ. Furthermore, an increase in the release of corticosterone triggered by T. cruzi-driven systemic inflammation is strongly associated with the alterations seen in the thymus of infected animals. Moreover, changes in the levels of other hormones, including growth hormone, prolactin, and testosterone are also able to contribute to the disruption of thymic homeostasis secondary to T. cruzi infection. In this review, we discuss the role of hormonal circuits involved in the normal T cell development and trafficking, as well as their role on the thymic alterations likely related to the peripheral T cell disturbances largely reported in both chagasic patients and animal models of Chagas disease.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Institute of Clinical and Experimental Immunology (IDICER-CONICET UNR), Rosario, Argentina
- *Correspondence: Ana Rosa Pérez, ,
| | - Alexandre Morrot
- Faculty of Medicine, Tuberculosis Research Center, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vinicius Frias Carvalho
- Laboratory of Inflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Carbajosa S, Gea S, Chillón-Marinas C, Poveda C, Del Carmen Maza M, Fresno M, Gironès N. Altered bone marrow lymphopoiesis and interleukin-6-dependent inhibition of thymocyte differentiation contribute to thymic atrophy during Trypanosoma cruzi infection. Oncotarget 2017; 8:17551-17561. [PMID: 28147332 PMCID: PMC5392268 DOI: 10.18632/oncotarget.14886] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 01/17/2017] [Indexed: 11/25/2022] Open
Abstract
Thymic atrophy occurs during infection being associated with apoptosis of double positive (DP) and premature exit of DP and double negative (DN) thymocytes. We observed for the first time that a significant bone marrow aplasia and a decrease in common lymphoid progenitors (CLPs) preceded thymic alterations in mice infected with Trypanosoma cruzi. In addition, depletion of the DN2 stage was previous to the DN1, indicating an alteration in the differentiation from DN1 to DN2 thymocytes. Interestingly, infected mice deficient in IL-6 expression showed higher numbers of DP and CD4+ thymocytes than wild type infected mice, while presenting similar percentages of DN1 thymocytes. Moreover, the drop in late differentiation stages of DN thymocytes was partially abrogated in comparison with wild type littermates. Thus, our results suggest that thymic atrophy involves a drop in CLPs production in bone marrow and IL-6-dependent and independent mechanisms that inhibits the differentiation of DN thymocytes.
Collapse
Affiliation(s)
- Sofía Carbajosa
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Susana Gea
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Carlos Chillón-Marinas
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Cristina Poveda
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - María Del Carmen Maza
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.,Instituto Sanitario de Investigación Princesa, Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.,Instituto Sanitario de Investigación Princesa, Madrid, Spain
| | - Núria Gironès
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.,Instituto Sanitario de Investigación Princesa, Madrid, Spain
| |
Collapse
|
19
|
Silva-dos-Santos D, Barreto-de-Albuquerque J, Guerra B, Moreira OC, Berbert LR, Ramos MT, Mascarenhas BAS, Britto C, Morrot A, Serra Villa-Verde DM, Garzoni LR, Savino W, Cotta-de-Almeida V, de Meis J. Unraveling Chagas disease transmission through the oral route: Gateways to Trypanosoma cruzi infection and target tissues. PLoS Negl Trop Dis 2017; 11:e0005507. [PMID: 28379959 PMCID: PMC5397068 DOI: 10.1371/journal.pntd.0005507] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 04/19/2017] [Accepted: 03/20/2017] [Indexed: 02/02/2023] Open
Abstract
Oral transmission of Trypanosoma cruzi, the causative agent of Chagas disease, is the most important route of infection in Brazilian Amazon and Venezuela. Other South American countries have also reported outbreaks associated with food consumption. A recent study showed the importance of parasite contact with oral cavity to induce a highly severe acute disease in mice. However, it remains uncertain the primary site of parasite entry and multiplication due to an oral infection. Here, we evaluated the presence of T. cruzi Dm28c luciferase (Dm28c-luc) parasites in orally infected mice, by bioluminescence and quantitative real-time PCR. In vivo bioluminescent images indicated the nasomaxillary region as the site of parasite invasion in the host, becoming consistently infected throughout the acute phase. At later moments, 7 and 21 days post-infection (dpi), luminescent signal is denser in the thorax, abdomen and genital region, because of parasite dissemination in different tissues. Ex vivo analysis demonstrated that the nasomaxillary region, heart, mandibular lymph nodes, liver, spleen, brain, epididymal fat associated to male sex organs, salivary glands, cheek muscle, mesenteric fat and lymph nodes, stomach, esophagus, small and large intestine are target tissues at latter moments of infection. In the same line, amastigote nests of Dm28c GFP T. cruzi were detected in the nasal cavity of 6 dpi mice. Parasite quantification by real-time qPCR at 7 and 21 dpi showed predominant T. cruzi detection and expansion in mouse nasal cavity. Moreover, T. cruzi DNA was also observed in the mandibular lymph nodes, pituitary gland, heart, liver, small intestine and spleen at 7 dpi, and further, disseminated to other tissues, such as the brain, stomach, esophagus and large intestine at 21 dpi. Our results clearly demonstrated that oral cavity and adjacent compartments is the main target region in oral T. cruzi infection leading to parasite multiplication at the nasal cavity. Oral transmission of Trypanosoma cruzi associated with food/beverage consumption is presently an important route of infection in Brazil and Venezuela. Colombia, Bolivia, Argentina and Ecuador have also reported to have acute cases of Chagas disease transmission through the oral route. Significant studies about this form of T. cruzi infection are largely lacking. In addition to the classic cardiac involvement, orally-infected patient progress to a highly symptomatic disease and increased mortality rate (8–35%), surpassing the calculated mortality produced by the disease resulting from the biting of infected insect vectors (5–10%). Here, we explored by in vivo bioluminescent images, qPCR and fluorescence microscopy the primary site of parasite entry and multiplication in oral infection (OI). Our results clearly demonstrated that the oral cavity is the main T. cruzi target region in OI, leading to parasite multiplication at the nasal cavity and parasite dissemination to the brain and peripheral tissues. Interestingly, facial edema, paraesthesia of the tongue, gingivitis and dry cough were already described in affected patients. These findings might be associated to our present data, which describe for the first time the nasomaxillary region as the main target tissue following oral T. cruzi infection.
Collapse
Affiliation(s)
- Danielle Silva-dos-Santos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Bárbara Guerra
- National Center of Structural Biology and Bio-imaging—CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Otacilio C. Moreira
- Laboratory on Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Luiz Ricardo Berbert
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Mariana Tavares Ramos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Constança Britto
- Laboratory on Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Alexandre Morrot
- Department of Immunology, Microbiology Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Déa M. Serra Villa-Verde
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Luciana Ribeiro Garzoni
- Laboratory for Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
20
|
Morrot A, Villar SR, González FB, Pérez AR. Evasion and Immuno-Endocrine Regulation in Parasite Infection: Two Sides of the Same Coin in Chagas Disease? Front Microbiol 2016; 7:704. [PMID: 27242726 PMCID: PMC4876113 DOI: 10.3389/fmicb.2016.00704] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/28/2016] [Indexed: 12/16/2022] Open
Abstract
Chagas disease is a serious illness caused by the protozoan parasite Trypanosoma cruzi. Nearly 30% of chronically infected people develop cardiac, digestive, or mixed alterations, suggesting a broad range of host-parasite interactions that finally impact upon chronic disease outcome. The ability of T. cruzi to persist and cause pathology seems to depend on diverse factors like T. cruzi strains, the infective load and the route of infection, presence of virulence factors, the parasite capacity to avoid protective immune response, the strength and type of host defense mechanisms and the genetic background of the host. The host-parasite interaction is subject to a constant neuro-endocrine regulation that is thought to influence the adaptive immune system, and as the infection proceeds it can lead to a broad range of outcomes, ranging from pathogen elimination to its continued persistence in the host. In this context, T. cruzi evasion strategies and host defense mechanisms can be envisioned as two sides of the same coin, influencing parasite persistence and different outcomes observed in Chagas disease. Understanding how T. cruzi evade host's innate and adaptive immune response will provide important clues to better dissect mechanisms underlying the pathophysiology of Chagas disease.
Collapse
Affiliation(s)
- Alexandre Morrot
- Institute of Microbiology, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Silvina R Villar
- Institute of Clinical and Experimental Immunology of Rosario, CONICET, National University of RosarioRosario, Argentina; Faculty of Medical Sciences, National University of RosarioRosario, Argentina
| | - Florencia B González
- Institute of Clinical and Experimental Immunology of Rosario, CONICET, National University of RosarioRosario, Argentina; Faculty of Medical Sciences, National University of RosarioRosario, Argentina
| | - Ana R Pérez
- Institute of Clinical and Experimental Immunology of Rosario, CONICET, National University of RosarioRosario, Argentina; Faculty of Medical Sciences, National University of RosarioRosario, Argentina
| |
Collapse
|
21
|
Domingues CS, Hardoim DJ, Souza CSF, Cardoso FO, Mendes VG, Previtalli-Silva H, Abreu-Silva AL, Pelajo-Machado M, Gonçalves da Costa SC, Calabrese KS. Oral Outbreak of Chagas Disease in Santa Catarina, Brazil: Experimental Evaluation of a Patient's Strain. PLoS One 2015; 10:e0122566. [PMID: 26469517 PMCID: PMC4607495 DOI: 10.1371/journal.pone.0122566] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/22/2015] [Indexed: 11/18/2022] Open
Abstract
Chagas disease is a worldwide public health problem. Although the vectorial transmission of Chagas disease has been controlled in Brazil there are other ways of transmission, such as the ingestion of T. cruzi contaminated food, which ensures the continuation of this zoonosis. Here, we demonstrate the influence of the inoculation route on the establishment and development of the SC2005 T. cruzi strain infection in mice. Groups of Swiss mice were infected intragastrically (IG) or intraperitoneally (IP) with the T. cruzi SC2005 strain derived from an outbreak of oral Chagas disease. The results revealed that 100% of IP infected mice showed parasitemia, while just 36% of IG infected showed the presence of the parasite in blood. The parasitemia peaks were later and less intense in the IG infected mice. Mortality of the IP infected animals was more intense and earlier when compared to the IG infected mice. In the IP infected mice leucopenia occurred in the early infection followed by leucocytosis, correlating positively with the increase of the parasites. However, in the IG infected mice only an increase in monocytes was observed, which was positively correlated with the increase of the parasites. Histopathological analyses revealed a myotropic pattern of the SC2005 strain with the presence of inflammatory infiltrates and parasites in different organs of the animals infected by both routes as well as fibrosis foci and collagen redistribution. The flow cytometric analysis demonstrated a fluctuation of the T lymphocyte population in the blood, spleen and mesenteric lymph nodes of the infected animals. T. cruzi DNA associated with the presence of inflammatory infiltrates was detected by PCR in the esophagus, stomach and intestine of all infected mice. These findings are important for the understanding of the pathogenesis of T. cruzi infection by both inoculation routes.
Collapse
Affiliation(s)
- Carolina S. Domingues
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Daiana J. Hardoim
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Celeste S. F. Souza
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Flávia O. Cardoso
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Verônica G. Mendes
- Centro de Desenvolvimento Tecnológico em Saúde (CDTS)/Instituto Nacional de Ciência e Tecnologia em Doenças Negligenciadas/FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Henrique Previtalli-Silva
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Ana L. Abreu-Silva
- Departamento de Patologia, Universidade Estadual do Maranhão, São Luiz, Maranhão, Brasil
| | - Marcelo Pelajo-Machado
- Laboratório de Patologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | | | - Kátia S. Calabrese
- Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brasil
- * E-mail:
| |
Collapse
|
22
|
Immune Evasion Strategies of Trypanosoma cruzi. J Immunol Res 2015; 2015:178947. [PMID: 26240832 PMCID: PMC4512591 DOI: 10.1155/2015/178947] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 12/31/2014] [Indexed: 01/03/2023] Open
Abstract
Microbes have evolved a diverse range of strategies to subvert the host immune system. The protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease, provides a good example of such adaptations. This parasite targets a broad spectrum of host tissues including both peripheral and central lymphoid tissues. Rapid colonization of the host gives rise to a systemic acute response which the parasite must overcome. The parasite in fact undermines both innate and adaptive immunity. It interferes with the antigen presenting function of dendritic cells via an action on host sialic acid-binding Ig-like lectin receptors. These receptors also induce suppression of CD4(+) T cells responses, and we presented evidence that the sialylation of parasite-derived mucins is required for the inhibitory effects on CD4 T cells. In this review we highlight the major mechanisms used by Trypanosoma cruzi to overcome host immunity and discuss the role of parasite colonization of the central thymic lymphoid tissue in chronic disease.
Collapse
|
23
|
Barreto-de-Albuquerque J, Silva-dos-Santos D, Pérez AR, Berbert LR, de Santana-van-Vliet E, Farias-de-Oliveira DA, Moreira OC, Roggero E, de Carvalho-Pinto CE, Jurberg J, Cotta-de-Almeida V, Bottasso O, Savino W, de Meis J. Trypanosoma cruzi Infection through the Oral Route Promotes a Severe Infection in Mice: New Disease Form from an Old Infection? PLoS Negl Trop Dis 2015; 9:e0003849. [PMID: 26090667 PMCID: PMC4474863 DOI: 10.1371/journal.pntd.0003849] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/26/2015] [Indexed: 11/16/2022] Open
Abstract
Oral transmission of Chagas disease has been documented in Latin American countries. Nevertheless, significant studies on the pathophysiology of this form of infection are largely lacking. The few studies investigating oral route infection disregard that inoculation in the oral cavity (Oral infection, OI) or by gavage (Gastrointestinal infection, GI) represent different infection routes, yet both show clear-cut parasitemia and heart parasitism during the acute infection. Herein, BALB/c mice were subjected to acute OI or GI infection using 5x104 culture-derived Trypanosoma cruzi trypomastigotes. OI mice displayed higher parasitemia and mortality rates than their GI counterparts. Heart histopathology showed larger areas of infiltration in the GI mice, whereas liver lesions were more severe in the OI animals, accompanied by higher Alanine Transaminase and Aspartate Transaminase serum contents. A differential cytokine pattern was also observed because OI mice presented higher pro-inflammatory cytokine (IFN-γ, TNF) serum levels than GI animals. Real-time PCR confirmed a higher TNF, IFN-γ, as well as IL-10 expression in the cardiac tissue from the OI group compared with GI. Conversely, TGF-β and IL-17 serum levels were greater in the GI animals. Immunolabeling revealed macrophages as the main tissue source of TNF in infected mice. The high mortality rate observed in the OI mice paralleled the TNF serum rise, with its inhibition by an anti-TNF treatment. Moreover, differences in susceptibility between GIversusOI mice were more clearly related to the host response than to the effect of gastric pH on parasites, since infection in magnesium hydroxide-treated mice showed similar results. Overall, the present study provides conclusive evidence that the initial site of parasite entrance critically affects host immune response and disease outcome. In light of the occurrence of oral Chagas disease outbreaks, our results raise important implications in terms of the current view of the natural disease course and host-parasite relationship. Chagas disease caused by the protozoan Trypanosoma cruzi is endemic in Latin America and a neglected tropical disease, which affects 6–7 million people worldwide. Currently, oral transmission is the most frequent pathway of infection in Brazil but also occurs in other endemic countries. This important infection route is underestimated and understudied. Here, we demonstrate that the site of parasite entrance, in the oral cavity (OI), as observed in natural infection, or directly to the gastrointestinal tract (GI), differentially affects the host-immune response and mortality. OI promotes a severe acute disease, elevated parasitemia and TNF mediated mortality. OI showed intense hepatitis and mild heart damage. Interestingly, GI mice presented mild disease, along with less circulating TNF and higher TGF-β and IL-17 serum contents. GI animals showed mild liver damage and intense heart inflammation. Our study is a pioneer work that analyzes the features of two distinct routes of oral infection. In addition, it provides new clues for Chagas pathology and stimulates background for the elucidation of disease features in orally exposed populations.
Collapse
Affiliation(s)
| | - Danielle Silva-dos-Santos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ana Rosa Pérez
- Immunology Institute, Faculty of Medical Science, National University of Rosario, Rosario, Argentina
| | - Luiz Ricardo Berbert
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | | | - Otacilio C. Moreira
- Laboratory on Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Eduardo Roggero
- Immunology Institute, Faculty of Medical Science, National University of Rosario, Rosario, Argentina
| | | | - José Jurberg
- National and International Laboratory on Triatomine Taxonomy, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Oscar Bottasso
- Immunology Institute, Faculty of Medical Science, National University of Rosario, Rosario, Argentina
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
24
|
Kannen V, de Oliveira EC, Motta BZ, Chaguri AJ, Brunaldi MO, Garcia SB. Trypanosomiasis-induced megacolon illustrates how myenteric neurons modulate the risk for colon cancer in rats and humans. PLoS Negl Trop Dis 2015; 9:e0003744. [PMID: 25884710 PMCID: PMC4401552 DOI: 10.1371/journal.pntd.0003744] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/07/2015] [Indexed: 01/05/2023] Open
Abstract
Background Trypanosomiasis induces a remarkable myenteric neuronal degeneration leading to megacolon. Very little is known about the risk for colon cancer in chagasic megacolon patients. To clarify whether chagasic megacolon impacts on colon carcinogenesis, we investigated the risk for colon cancer in Trypanosoma cruzi (T. cruzi) infected patients and rats. Methods Colon samples from T. cruzi-infected and uninfected patients and rats were histopathologically investigated with colon cancer biomarkers. An experimental model for chemical myenteric denervation was also performed to verify the myenteric neuronal effects on colon carcinogenesis. All experiments complied the guidelines and approval of ethical institutional review boards. Results No colon tumors were found in chagasic megacolon samples. A significant myenteric neuronal denervation was observed. Epithelial cell proliferation and hyperplasia were found increased in chagasic megacolon. Analyzing the argyrophilic nucleolar organiser regions within the cryptal bottom revealed reduced risk for colon cancer in Chagas’ megacolon patients. T. cruzi-infected rats showed a significant myenteric neuronal denervation and decreased numbers of colon preneoplastic lesions. In chemical myenteric denervated rats preneoplastic lesions were reduced from the 2nd wk onward, which ensued having the colon myenteric denervation significantly induced. Conclusion/Significance Our data suggest that the trypanosomiasis-related myenteric neuronal degeneration protects the colon tissue from carcinogenic events. Current findings highlight potential mechanisms in tropical diseases and cancer research. The myenteric neuronal activity on colon carcinogenesis is a matter of debate. Chagas disease (a trypanosomiasis-related chronic infection) induces megacolon damaging myenteric neurons. Puzzling, tumors have been rarely reported in chagasic megacolon patients. We reveal here hyperplasia-related high-proliferation occurs in chagasic megacolon, although the risk for colon cancer is reduced. Having carcinogen-exposed rats infected with Trypanosoma cruzi reduced the numbers of myenteric neurons and colon preneoplastic lesions. An experimental model for chemical myenteric denervation was applied in carcinogen-exposed rats revealing that myenteric neurons promote the development of colon preneoplastic lesions. Yet, activity of the fecal content had to be secluded from the myenteric neuronal activity on colon carcinogenesis. Hartmann’s surgical procedure enabled that. This was applied together with carcinogenic exposure and myenteric neuronal denervation ensuring that the neuronal activity is associated with enhanced development of colon carcinogenesis. Taken together, we believe colon tumors are not found within the chagasic megacolon region because the myenteric neuronal density is impaired. These observations shed lights on novel potential cell to cell interactions promoting the colon cancer development.
Collapse
Affiliation(s)
- Vinicius Kannen
- Department of Pathology, University of Sao Paulo, Ribeirao Preto, Brazil
- * E-mail:
| | - Enio C. de Oliveira
- Department of Surgery, Medical School, Federal University of Goias, Goiania, Brazil
| | - Bruno Zene Motta
- Department of Pathology, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | | | - Sérgio B. Garcia
- Department of Pathology, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
25
|
Mendlovic F, Cruz-Rivera M, Ávila G, Vaughan G, Flisser A. Cytokine, antibody and proliferative cellular responses elicited by Taenia solium calreticulin upon experimental infection in hamsters. PLoS One 2015; 10:e0121321. [PMID: 25811778 PMCID: PMC4374884 DOI: 10.1371/journal.pone.0121321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/30/2015] [Indexed: 12/11/2022] Open
Abstract
Taenia solium causes two diseases in humans, cysticercosis and taeniosis. Tapeworm carriers are the main risk factor for neurocysticercosis. Limited information is available about the immune response elicited by the adult parasite, particularly the induction of Th2 responses, frequently associated to helminth infections. Calreticulin is a ubiquitous, multifunctional protein involved in cellular calcium homeostasis, which has been suggested to play a role in the regulation of immune responses. In this work, we assessed the effect of recombinant T. solium calreticulin (rTsCRT) on the cytokine, humoral and cellular responses upon experimental infection in Syrian Golden hamsters (Mesocricetus auratus). Animals were infected with T. solium cysticerci and euthanized at different times after infection. Specific serum antibodies, proliferative responses in mesenteric lymph nodes and spleen cells, as well as cytokines messenger RNA (mRNA) were analyzed. The results showed that one third of the infected animals elicited anti-rTsCRT IgG antibodies. Interestingly, mesenteric lymph node (MLN) cells from either infected or non-infected animals did not proliferate upon in vitro stimulation with rTsCRT. Additionally, stimulation with a tapeworm crude extract resulted in increased expression of IL-4 and IL-5 mRNA. Upon stimulation, rTsCRT increased the expression levels of IL-10 in spleen and MLN cells from uninfected and infected hamsters. The results showed that rTsCRT favors a Th2-biased immune response characterized by the induction of IL-10 in mucosal and systemic lymphoid organs. Here we provide the first data on the cytokine, antibody and cellular responses to rTsCRT upon in vitro stimulation during taeniasis.
Collapse
Affiliation(s)
- Fela Mendlovic
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, México D.F. 04510, México
- Facultad de Ciencias de la Salud, Universidad Anáhuac, México Norte, Av. Universidad Anáhuac 46, Huixquilucan, 52786 Edo. de México, México
| | - Mayra Cruz-Rivera
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, México D.F. 04510, México
| | - Guillermina Ávila
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, México D.F. 04510, México
| | | | - Ana Flisser
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán, México D.F. 04510, México
- * E-mail:
| |
Collapse
|
26
|
Trypanosoma cruzi trans-sialidase prevents elicitation of Th1 cell response via interleukin 10 and downregulates Th1 effector cells. Infect Immun 2015; 83:2099-108. [PMID: 25754197 DOI: 10.1128/iai.00031-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/01/2015] [Indexed: 12/22/2022] Open
Abstract
The trans-sialidases (TSs) from Trypanosoma cruzi, the agent of Chagas disease, are virulence factors shed to the bloodstream that induce strong alterations in the immune system. Here, we report that both enzymatically active TS (aTS) and its lectinlike isoform (iTS) disturb CD4 T cell physiology, inducing downregulation of Th1 cell functionality and in vivo cell expansion. By using ovalbumin-specific DO11.10 cells as tracers of clones developing the Th1 phenotype, we found that the infection induced significant amounts of gamma interferon (IFN-γ) but low levels of interleukin 2 (IL-2) and increased IL-4 production in vivo, in agreement with a mixed T helper response. The production of cytokines associated with the Th2 phenotype was prevented by passive transfer of anti-TS neutralizing antibodies. TSs also reduced the T cell receptor signaling as assayed by Zap-70 phosphorylation. TSs also reduced IL-2 and IFN-γ secretion, with a concomitant increase in IL-4 production and then an unbalancing of the CD4 T cell response toward the Th2 phenotype. This effect was prevented by using anti-IL-10 neutralizing antibodies or IL-10(-/-) antigen-presenting cells, supporting the subversion of this regulatory pathway. In support, TSs stimulated IL-10 secretion by antigen-presenting cells during their interaction with CD4 T cells. When polarized cells were stimulated in the presence of TSs, the secretion of IL-2 and IFN-γ was strongly downregulated in Th1 cells, while IL-2 production was upregulated in Th2 cells. Although the Th1 response is associated with host survival, it may simultaneously induce extensive damage to infected tissues. Thus, by delaying the elicitation of the Th1 response and limiting its effector properties, TSs restrain the cell response, supporting T. cruzi colonization and persistence while favoring host survival.
Collapse
|
27
|
de Meis J, Savino W. Mature peripheral T cells are important to preserve thymus function and selection of thymocytes during Mycobacterium tuberculosis infection. Immunotherapy 2014; 5:573-6. [PMID: 23725281 DOI: 10.2217/imt.13.41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Evaluation of: Nobrega C, Nunes-Alves C, Cerqueira-Rodrigues B et al. T cells home to the thymus and control infection. J. Immunol. 190, 1646-1658 (2013). It is well documented that the thymus is a target organ for a large variety of pathogens (virus, bacteria, fungi and protozoa). Moreover, the presence of pathogen-derived antigens in the thymus of infected mice seems to interfere with the capacity of mature T cells to respond to the invading organism. In this way, Nobrega and colleagues demonstrated in 2010 that Mycobacterium avium infection in the thymus leads to the appearance of differentiated T cells tolerogenic for bacterial antigens. In the present and elegant study, the same group demonstrates that T-cell recirculation from the periphery to the thymus is a mechanism that allows the immune system to respond to thymic infection. A Mycobacterium-infected thymus increases the production of Th1-effector chemokines, such as CXCL9 and CXCL10, which in turn recruit CXCR3(+) peripheral T cells involved in intrathymic bacterial control. Taken together, these findings may represent an important issue of the host response, in terms of different pathogens able to infect the thymus.
Collapse
Affiliation(s)
- Juliana de Meis
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Manguinhos, Rio de Janeiro, Brazil
| | | |
Collapse
|
28
|
Morrot A. The Role of Sialic Acid-Binding Receptors (Siglecs) in the Immunomodulatory Effects of Trypanosoma cruzi Sialoglycoproteins on the Protective Immunity of the Host. SCIENTIFICA 2013; 2013:965856. [PMID: 24455435 PMCID: PMC3885277 DOI: 10.1155/2013/965856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 12/10/2013] [Indexed: 06/03/2023]
Abstract
Chagas disease is caused by the protozoan parasite Trypanosoma cruzi and is an important endemic infection in Latin America. Lately, it has also become a health concern in the United States and Europe. Most of the immunomodulatory mechanisms associated with this parasitic infection have been attributed to mucin-like molecules on the T. cruzi surface. Mucins are high molecular weight glycoproteins that are involved in regulating diverse cellular activities in both normal and pathological conditions. In Trypanosoma cruzi infection, the parasite-derived mucins are the main acceptors of sialic acid and it has been suggested that they play a role in various host-parasite interactions during the course of Chagas disease. Recently, we have presented evidence that sialylation of the mucins is required for the inhibitory effects on CD4(+) T cells. In what follows we propose that signaling via sialic acid-binding Ig-like lectin receptors for these highly sialylated structures on host cells contributes to the arrest of cell cycle progression in the G1 phase and may allow the parasite to modulate the immune system of the host.
Collapse
Affiliation(s)
- Alexandre Morrot
- Institute of Microbiology, Federal University of Rio de Janeiro, CCS, Sala D1-035, Avenida Carlos Chagas Filho 373, Cidade Universitária, Ilha do Fundão, 21.941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
29
|
de Meis J, Barreto de Albuquerque J, Silva Dos Santos D, Farias-de-Oliveira DA, Berbert LR, Cotta-de-Almeida V, Savino W. Trypanosoma cruzi Entrance through Systemic or Mucosal Infection Sites Differentially Modulates Regional Immune Response Following Acute Infection in Mice. Front Immunol 2013; 4:216. [PMID: 23898334 PMCID: PMC3724200 DOI: 10.3389/fimmu.2013.00216] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/13/2013] [Indexed: 12/11/2022] Open
Abstract
Acute Chagas disease is characterized by a systemic infection that leads to the strong activation of the adaptive immune response. Outbreaks of oral contamination by the infective protozoan Trypanosoma cruzi are frequent in Brazil and other Latin American countries, and an increased severity of clinical manifestations and mortality is observed in infected patients. These findings have elicited questions about the specific responses triggered after T. cruzi entry via mucosal sites, possibly modulating local immune mechanisms, and further impacting regional and systemic immunity. Here, we provide evidence for the existence of differential lymphoid organ responses in experimental models of acute T. cruzi infection.
Collapse
Affiliation(s)
- Juliana de Meis
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | | | | | | | | | | | | |
Collapse
|
30
|
Sanoja C, Carbajosa S, Fresno M, Gironès N. Analysis of the dynamics of infiltrating CD4(+) T cell subsets in the heart during experimental Trypanosoma cruzi infection. PLoS One 2013; 8:e65820. [PMID: 23776551 PMCID: PMC3679147 DOI: 10.1371/journal.pone.0065820] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 04/28/2013] [Indexed: 01/25/2023] Open
Abstract
Chagas disease, caused by the protozoan parasite Trypanosoma cruzi, affects several million people in Latin America. Myocarditis, observed during both the acute and chronic phases of the disease, is characterized by an inflammatory mononuclear cell infiltrate that includes CD4+ T cells. It is known that Th1 cytokines help to control infection. The role that Treg and Th17 cells may play in disease outcome, however, has not been completely elucidated. We performed a comparative study of the dynamics of CD4+ T cell subsets after infection with the T. cruzi Y strain during both the acute and chronic phases of the disease using susceptible BALB/c and non-susceptible C57BL/6 mice infected with high or low parasite inocula. During the acute phase, infected C57BL/6 mice showed high levels of CD4+ T cell infiltration and expression of Th1 cytokines in the heart associated with the presence of Treg cells. In contrast, infected BALB/c mice had a high heart parasite burden, low heart CD4+ T cell infiltration and low levels of Th1 and inflammatory cytokines, but with an increased presence of Th17 cells. Moreover, an increase in the expression of IL-6 in susceptible mice was associated with lethality upon infection with a high parasite load. Chronically infected BALB/c mice continued to present higher parasite burdens than C57BL/6 mice and also higher levels of IFN-γ, TNF, IL-10 and TGF-β. Thus, the regulation of the Th1 response by Treg cells in the acute phase may play a protective role in non-susceptible mice irrespective of parasite numbers. On the other hand, Th17 cells may protect susceptible mice at low levels of infection, but could, in association with IL-6, be pathogenic at high parasite loads.
Collapse
Affiliation(s)
- Cristina Sanoja
- Instituto de Biología Experimental, Universidad Central de Venezuela (UCV), Caracas, Venezuela
| | - Sofía Carbajosa
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)–Universidad Autónoma de Madrid (UAM), Cantoblanco, Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)–Universidad Autónoma de Madrid (UAM), Cantoblanco, Madrid, Spain
- Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Núria Gironès
- Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas (CSIC)–Universidad Autónoma de Madrid (UAM), Cantoblanco, Madrid, Spain
- Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
- * E-mail:
| |
Collapse
|
31
|
Deobagkar-Lele M, Chacko SK, Victor ES, Kadthur JC, Nandi D. Interferon-γ- and glucocorticoid-mediated pathways synergize to enhance death of CD4(+) CD8(+) thymocytes during Salmonella enterica serovar Typhimurium infection. Immunology 2013. [PMID: 23186527 DOI: 10.1111/imm.12047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Thymic atrophy is known to occur during infections; however, there is limited understanding of its causes and of the cross-talk between different pathways. This study investigates mechanisms involved in thymic atrophy during a model of oral infection by Salmonella enterica serovar Typhimurium (S. typhimurium). Significant death of CD4(+) CD8(+) thymocytes, but not of single-positive thymocytes or peripheral lymphocytes, is observed at later stages during infection with live, but not heat-killed, bacteria. The death of CD4(+) CD8(+) thymocytes is Fas-independent as shown by infection studies with lpr mice. However, apoptosis occurs with lowering of mitochondrial potential and higher caspase-3 activity. The amounts of cortisol, a glucocorticoid, and interferon-γ (IFN-γ), an inflammatory cytokine, increase upon infection. To investigate the functional roles of these molecules, studies were performed using Ifnγ(-/-) mice together with RU486, a glucocorticoid receptor antagonist. Treatment of C57BL/6 mice with RU486 does not affect colony-forming units (CFU), amounts of IFN-γ and mouse survival; however, there is partial rescue in thymocyte death. Upon infection, Ifnγ(-/-) mice display higher CFU and lower survival but more surviving thymocytes are recovered. However, there is no difference in cortisol amounts in C57BL/6 and Ifnγ(-/-) mice. Importantly, the number of CD4(+) CD8(+) thymocytes is significantly higher in Ifnγ(-/-) mice treated with RU486 along with lower caspase-3 activity and mitochondrial damage. Hence, endogenous glucocorticoid and IFN-γ-mediated pathways are parallel but synergize in an additive manner to induce death of CD4(+) CD8(+) thymocytes during S. typhimurium infection. The implications of this study for host responses during infection are discussed.
Collapse
|
32
|
Nardy AFFR, Luiz da Silva Filho J, Pérez AR, de Meis J, Farias-de-Oliveira DA, Penha L, de Araújo Oliveira I, Dias WB, Todeschini AR, Freire-de-Lima CG, Bellio M, Caruso-Neves C, Pinheiro AA, Takiya CM, Bottasso O, Savino W, Morrot A. Trans-sialidase from Trypanosoma cruzi enhances the adhesion properties and fibronectin-driven migration of thymocytes. Microbes Infect 2013; 15:365-74. [PMID: 23481510 DOI: 10.1016/j.micinf.2013.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/28/2013] [Accepted: 02/08/2013] [Indexed: 01/06/2023]
Abstract
In experimental Trypanosoma cruzi infections, severe thymic atrophy leads to release of activated CD4(+)CD8(+) double-positive (DP) T cells to the periphery. In humans, activated DP T cells are found in the blood in association with severe cardiac forms of human chronic Chagas disease. The mechanisms underlying the premature thymocyte release during the chagasic thymic atrophy remain elusive. We tested whether the migratory properties of intrathymic thymocytes are modulated by the parasite trans-sialidase (TS). We found that TS affected the dynamics of thymocytes undergoing intrathymic maturation, and these changes were accompanied by an increase in the number of recent DP thymic emigrants in the peripheral lymphoid organs. We demonstrated that increased percentages of blood DP T cell subsets were associated with augmented antibody titers against TS in chagasic patients with chronic cardiomyopathy. In vitro studies showed that TS was able to activate the MAPK pathway and actin filament mobilization in thymocytes. These effects were correlated with its ability to modulate the adhesion of thymocytes to thymic epithelial cells and their migration toward extracellular matrix. These findings point to effects of TS that could influence the escape of immature thymocytes in Chagas disease.
Collapse
Affiliation(s)
- Ana Flávia F R Nardy
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Basso B. Modulation of immune response in experimental Chagas disease. World J Exp Med 2013; 3:1-10. [PMID: 24520540 PMCID: PMC3905588 DOI: 10.5493/wjem.v3.i1.1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/07/2013] [Accepted: 02/06/2013] [Indexed: 02/06/2023] Open
Abstract
Trypanosoma cruzi (T. cruzi), the etiological agent of Chagas disease, affects nearly 18 million people in Latin America and 90 million are at risk of infection. The parasite presents two stages of medical importance in the host, the amastigote, intracellular replicating form, and the extracellular trypomastigote, the infective form. Thus infection by T. cruzi induces a complex immune response that involves effectors and regulatory mechanisms. That is why control of the infection requires a strong humoral and cellular immune response; hence, the outcome of host-parasite interaction in the early stages of infection is extremely important. A critical event during this period of the infection is innate immune response, in which the macrophage’s role is vital. Thus, after being phagocytized, the parasite is able to develop intracellularly; however, during later periods, these cells induce its elimination by means of toxic metabolites. In turn, as the infection progresses, adaptive immune response mechanisms are triggered through the TH1 and TH2 responses. Finally, T. cruzi, like other protozoa such as Leishmania and Toxoplasma, have numerous evasive mechanisms to the immune response that make it possible to spread around the host. In our Laboratory we have developed a vaccination model in mice with Trypanosoma rangeli, nonpathogenic to humans, which modulates the immune response to infection by T. cruzi, thus protecting them. Vaccinated animals showed an important innate response (modulation of NO and other metabolites, cytokines, activation of macrophages), a strong adaptive cellular response and significant increase in specific antibodies. The modulation caused early elimination of the parasites, low parasitaemia, the absence of histological lesions and high survival rates. Even though progress has been made in the knowledge of some of these mechanisms, new studies must be conducted which could target further prophylactic and therapeutic trials against T. cruzi infection.
Collapse
|
34
|
Calderón J, Maganto-Garcia E, Punzón C, Carrión J, Terhorst C, Fresno M. The receptor Slamf1 on the surface of myeloid lineage cells controls susceptibility to infection by Trypanosoma cruzi. PLoS Pathog 2012; 8:e1002799. [PMID: 22807679 PMCID: PMC3395606 DOI: 10.1371/journal.ppat.1002799] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 05/30/2012] [Indexed: 02/06/2023] Open
Abstract
Trypanosoma cruzi, the protozoan parasite responsible for Chagas' disease, causes severe myocarditis often resulting in death. Here, we report that Slamf1-/- mice, which lack the hematopoietic cell surface receptor Slamf1, are completely protected from an acute lethal parasite challenge. Cardiac damage was reduced in Slamf1-/- mice compared to wild type mice, infected with the same doses of parasites, as a result of a decrease of the number of parasites in the heart even the parasitemia was only marginally less. Both in vivo and in vitro experiments reveal that Slamf1-defIcient myeloid cells are impaired in their ability to replicate the parasite and show altered production of cytokines. Importantly, IFN-γ production in the heart of Slamf1 deficient mice was much lower than in the heart of wt mice even though the number of infiltrating dendritic cells, macrophages, CD4 and CD8 T lymphocytes were comparable. Administration of an anti-Slamf1 monoclonal antibody also reduced the number of parasites and IFN-γ in the heart. These observations not only explain the reduced susceptibility to in vivo infection by the parasite, but they also suggest human Slamf1 as a potential target for therapeutic target against T. cruzi infection.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Antibodies, Protozoan/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/physiology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Chagas Cardiomyopathy/immunology
- Chagas Cardiomyopathy/parasitology
- Chagas Disease/immunology
- Chagas Disease/parasitology
- Cytokines/biosynthesis
- Dendritic Cells/immunology
- Disease Susceptibility
- Heart/parasitology
- Interferon-gamma/biosynthesis
- Macrophages/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Myeloid Cells/metabolism
- Myeloid Cells/parasitology
- Myocardium/metabolism
- Parasitemia
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Signaling Lymphocytic Activation Molecule Family Member 1
- Trypanosoma cruzi/immunology
- Trypanosoma cruzi/physiology
Collapse
Affiliation(s)
- Jossela Calderón
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Elena Maganto-Garcia
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Carmen Punzón
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - Javier Carrión
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Cox Terhorst
- The Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Manuel Fresno
- Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
- * E-mail:
| |
Collapse
|
35
|
Different infective forms trigger distinct immune response in experimental Chagas disease. PLoS One 2012; 7:e32912. [PMID: 22412949 PMCID: PMC3296760 DOI: 10.1371/journal.pone.0032912] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Accepted: 02/04/2012] [Indexed: 11/26/2022] Open
Abstract
Although metacyclic and blood trypomastigotes are completely functional in relation to parasite-host interaction and/or target cell invasion, they differ in the molecules present on the surface. Thus, aspects related to the variability that the forms of T. cruzi interacts with host cells may lead to fundamental implications on the immune response against this parasite and, consequently, the clinical evolution of Chagas disease. We have shown that BT infected mice presented higher levels of parasitemia during all the acute phase of infection. Moreover, the infection with either MT or BT forms resulted in increased levels of total leukocytes, monocytes and lymphocytes, specifically later for MT and earlier for BT. The infection with BT forms presented earlier production of proinflammatory cytokine TNF-α and later of IFN-γ by both T cells subpopulations. This event was accompanied by an early cardiac inflammation with an exacerbation of this process at the end of the acute phase. On the other hand, infection with MT forms result in an early production of IFN-γ, with subsequent control in the production of this cytokine by IL-10, which provided to these animals an immunomodulatory profile in the end of the acute phase. These results are in agreement with what was found for cardiac inflammation where animals infected with MT forms showed intense cardiac inflammation later at infection, with a decrease in the same at the end of this phase. In summary, our findings emphasize the importance of taking into account the inoculums source of T. cruzi, since vectorial or transfusional routes of T. cruzi infection may trigger distinct parasite-host interactions during the acute phase that may influence relevant biological aspects of chronic Chagas disease.
Collapse
|
36
|
Dynamics of Lymphocyte Populations during Trypanosoma cruzi Infection: From Thymocyte Depletion to Differential Cell Expansion/Contraction in Peripheral Lymphoid Organs. J Trop Med 2012; 2012:747185. [PMID: 22505943 PMCID: PMC3306984 DOI: 10.1155/2012/747185] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 10/17/2011] [Indexed: 01/05/2023] Open
Abstract
The comprehension of the immune responses in infectious diseases is crucial for developing novel therapeutic strategies. Here, we review current findings on the dynamics of lymphocyte subpopulations following experimental acute infection by Trypanosoma cruzi, the causative agent of Chagas disease. In the thymus, although the negative selection process of the T-cell repertoire remains operational, there is a massive thymocyte depletion and abnormal release of immature CD4+CD8+ cells to peripheral lymphoid organs, where they acquire an activated phenotype similar to activated effector or memory T cells. These cells apparently bypassed the negative selection process, and some of them are potentially autoimmune. In infected animals, an atrophy of mesenteric lymph nodes is also observed, in contrast with the lymphocyte expansion in spleen and subcutaneous lymph nodes, illustrating a complex and organ specific dynamics of lymphocyte subpopulations. Accordingly, T- and B-cell activation is seen in subcutaneous lymph nodes and spleen, but not in mesenteric lymph nodes. Lastly, although the function of peripheral CD4+CD8+ T-cell population remains to be defined in vivo, their presence may contribute to the immunopathological events found in both murine and human Chagas disease.
Collapse
|
37
|
Thymus atrophy and double-positive escape are common features in infectious diseases. J Parasitol Res 2012; 2012:574020. [PMID: 22518275 PMCID: PMC3307005 DOI: 10.1155/2012/574020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 10/20/2011] [Indexed: 11/21/2022] Open
Abstract
The thymus is a primary lymphoid organ in which bone marrow-derived T-cell precursors undergo differentiation, leading to migration of positively selected thymocytes to the T-cell-dependent areas of secondary lymphoid organs. This organ can undergo atrophy, caused by several endogenous and exogenous factors such as ageing, hormone fluctuations, and infectious agents. This paper will focus on emerging data on the thymic atrophy caused by infectious agents. We present data on the dynamics of thymus lymphocytes during acute Trypanosoma cruzi infection, showing that the resulting thymus atrophy comprises the abnormal release of thymic-derived T cells and may have an impact on host immune response.
Collapse
|
38
|
Dhiman M, Garg NJ. NADPH oxidase inhibition ameliorates Trypanosoma cruzi-induced myocarditis during Chagas disease. J Pathol 2011; 225:583-96. [PMID: 21952987 DOI: 10.1002/path.2975] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 07/04/2011] [Accepted: 07/18/2011] [Indexed: 12/15/2022]
Abstract
Trypanosoma cruzi, the aetiological agent of Chagas disease, invades nucleated mammalian cells including macrophages. In this study, we investigated the crosstalk between T. cruzi-induced immune activation of reactive oxygen species (ROS) and pro-inflammatory responses, and their role in myocardial pathology. Splenocytes of infected mice (C3H/HeN) responded to Tc-antigenic stimulus by more than a two-fold increase in NADPH oxidase (NOX) activity, ROS generation, cytokine production (IFN-γ > IL-4 > TNFα > IL1-β≈ IL6), and predominant expansion of CD4(+) and CD8(+) T cells. Inhibition of NOX, but not of myeloperoxidase and xanthine oxidase, controlled the ROS (>98%) and cytokine (70-89%) release by Tc-stimulated splenocytes of infected mice. Treatment of infected mice with apocynin (NOX inhibitor) in drinking water resulted in a 50-90% decline in endogenous NOX/ROS and cytokine levels, and splenic phagocytes' proliferation. The splenic percentage of T cells was maintained, though more than a 40% decline in splenic index (spleen weight/body weight) indicated decreased T-cell proliferation in apocynin-treated/infected mice. The blood and tissue parasite burden were significantly increased in apocynin-treated/infected mice, yet acute myocarditis, ie inflammatory infiltrate consisting of macrophages, neutrophils, and CD8(+) T cells, and tissue oxidative adducts (eg 8-isoprostanes, 3-nitrotyrosine, and 4-hydroxynonenal) were diminished in apocynin-treated/infected mice. Consequently, hypertrophy (increased cardiomyocytes' size and β-MHC, BNP, and ANP mRNA levels) and fibrosis (increased collagen, glycosaminoglycans, and lipid contents) of the heart during the chronic phase were controlled in apocynin-treated mice. We conclude that NOX/ROS is a critical regulator of the splenic response (phagocytes, T cells, and cytokines) to T. cruzi infection, and bystander effects of heart-infiltrating phagocytes and CD8(+) T cells resulting in cardiac remodelling in chagasic mice.
Collapse
Affiliation(s)
- Monisha Dhiman
- Department of Microbiology and Immunology, Center for Tropical Diseases, University of Texas Medical Branch, Galveston, TX 77555-1070, USA
| | | |
Collapse
|
39
|
Flores-García Y, Rosales-Encina JL, Satoskar AR, Talamás-Rohana P. IL-10-IFN-γ double producers CD4+ T cells are induced by immunization with an amastigote stage specific derived recombinant protein of Trypanosoma cruzi. Int J Biol Sci 2011; 7:1093-100. [PMID: 21927578 PMCID: PMC3174386 DOI: 10.7150/ijbs.7.1093] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2011] [Accepted: 07/20/2011] [Indexed: 12/30/2022] Open
Abstract
During the acute phase of infection, T. cruzi replicates extensively and releases immunomodulatory molecules that delay parasite-specific responses mediated by effector T cells. This mechanism of evasion allows the parasite to spread in the host. Parasite molecules that regulate the host immune response during Chagas'disease have not been fully identified. GPI-anchored mucins, glycoinositolphospholipids, and glycoproteins comprise some of the most abundant T. cruzi surface molecules. IL-10 IFN-γ-secreting CD4+ T cells are activated during chronic infections and are responsible for prolonged persistence of parasite and for host protection against severe inflammatory responses. In this work we evaluated the role of rMBP::SSP4 protein of T. cruzi, a recombinant protein derived from a GPI anchored antigen, SSP4, as an immunomodulator molecule, finding that it was able to induce high concentrations of IL-10 and IFN-γ both in vivo and in vitro; during this last condition, both cytokines were produced by IL-10-IFN-γ-secreting CD4+ T cells.
Collapse
Affiliation(s)
- Yevel Flores-García
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México, DF. 07360, Mexico
| | | | | | | |
Collapse
|
40
|
Morrot A, Terra-Granado E, Pérez AR, Silva-Barbosa SD, Milićević NM, Farias-de-Oliveira DA, Berbert LR, De Meis J, Takiya CM, Beloscar J, Wang X, Kont V, Peterson P, Bottasso O, Savino W. Chagasic thymic atrophy does not affect negative selection but results in the export of activated CD4+CD8+ T cells in severe forms of human disease. PLoS Negl Trop Dis 2011; 5:e1268. [PMID: 21858238 PMCID: PMC3156684 DOI: 10.1371/journal.pntd.0001268] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 06/21/2011] [Indexed: 11/19/2022] Open
Abstract
Extrathymic CD4+CD8+ double-positive (DP) T cells are increased in some pathophysiological conditions, including infectious diseases. In the murine model of Chagas disease, it has been shown that the protozoan parasite Trypanosoma cruzi is able to target the thymus and induce alterations of the thymic microenvironment and the lymphoid compartment. In the acute phase, this results in a severe atrophy of the organ and early release of DP cells into the periphery. To date, the effect of the changes promoted by the parasite infection on thymic central tolerance has remained elusive. Herein we show that the intrathymic key elements that are necessary to promote the negative selection of thymocytes undergoing maturation during the thymopoiesis remains functional during the acute chagasic thymic atrophy. Intrathymic expression of the autoimmune regulator factor (Aire) and tissue-restricted antigen (TRA) genes is normal. In addition, the expression of the proapoptotic Bim protein in thymocytes was not changed, revealing that the parasite infection-induced thymus atrophy has no effect on these marker genes necessary to promote clonal deletion of T cells. In a chicken egg ovalbumin (OVA)-specific T-cell receptor (TCR) transgenic system, the administration of OVA peptide into infected mice with thymic atrophy promoted OVA-specific thymocyte apoptosis, further indicating normal negative selection process during the infection. Yet, although the intrathymic checkpoints necessary for thymic negative selection are present in the acute phase of Chagas disease, we found that the DP cells released into the periphery acquire an activated phenotype similar to what is described for activated effector or memory single-positive T cells. Most interestingly, we also demonstrate that increased percentages of peripheral blood subset of DP cells exhibiting an activated HLA-DR+ phenotype are associated with severe cardiac forms of human chronic Chagas disease. These cells may contribute to the immunopathological events seen in the Chagas disease. The thymus is a primary lymphoid organ that plays an important role on the development of the immune system and maturation of the T cell repertoire. During the normal life span, this organ undergoes involution during the aging and also in the presence of a wide variety of infectious diseases. It has been shown that the protozoan parasite Trypanosoma cruzi is able to target the thymus and induce alterations of the thymic microenvironment. In the acute phase, this results in a severe atrophy of the organ and early release of immature double-positive (DP) T cells into the periphery. The effect of the changes promoted by the parasite infection on thymic central tolerance has remained not clear. The present study shows that the intrathymic key elements that promote the negative selection of thymocytes during the thymopoiesis remains functional in the acute chagasic thymic atrophy. However, we found that the DP cells released into the periphery acquire an activated phenotype and its high frequency in the peripheral blood are associated with severe cardiac forms of human chronic Chagas disease.
Collapse
Affiliation(s)
- Alexandre Morrot
- Department of Immunology, Microbiology Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Marini V, Moretti E, Bermejo D, Basso B. Vaccination with Trypanosoma rangeli modulates the profiles of immunoglobulins and IL-6 at local and systemic levels in the early phase of Trypanosoma cruzi experimental infection. Mem Inst Oswaldo Cruz 2011; 106:32-7. [PMID: 21340352 DOI: 10.1590/s0074-02762011000100005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 12/08/2010] [Indexed: 12/17/2022] Open
Abstract
In America, there are two species of Trypanosoma that can infect humans: Trypanosoma cruzi, which is responsible for Chagas disease and Trypanosoma rangeli, which is not pathogenic. We have developed a model of vaccination in mice with T. rangeli epimastigotes that protects against T. cruzi infection. The goal of this work was to study the pattern of specific immunoglobulins in the peritoneum (the site of infection) and in the sera of mice immunized with T. rangeli before and after challenge with T. cruzi. Additionally, we studied the effects triggered by antigen-antibodies binding and the levels of key cytokines involved in the humoral response, such as IL-4, IL-5 and IL-6. The immunization triggered the production of antibodies reactive with T. cruzi in peritoneal fluid (PF) and in serum, mainly IgG1 and, to a lesser magnitude, IgG2. Only immunized mice developed specific IgG3 antibodies in their peritoneal cavities. Antibodies were able to bind to the surface of the parasites and agglutinate them. Among the cytokines studied, IL-6 was elevated in PF during early infection, with higher levels in non-immunized-infected mice. The results indicate that T. rangeli vaccination against T. cruzi infection triggers a high production of specific IgG isotypes in PF and sera before infection and modulates the levels of IL-6 in PF in the early periods of infection.
Collapse
Affiliation(s)
- Vanina Marini
- Laboratorio de la Coordinación Nacional de Control de Vectores, Córdoba, Argentina
| | | | | | | |
Collapse
|
42
|
Teixeira ARL, Hecht MM, Guimaro MC, Sousa AO, Nitz N. Pathogenesis of chagas' disease: parasite persistence and autoimmunity. Clin Microbiol Rev 2011; 24:592-630. [PMID: 21734249 PMCID: PMC3131057 DOI: 10.1128/cmr.00063-10] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Acute Trypanosoma cruzi infections can be asymptomatic, but chronically infected individuals can die of Chagas' disease. The transfer of the parasite mitochondrial kinetoplast DNA (kDNA) minicircle to the genome of chagasic patients can explain the pathogenesis of the disease; in cases of Chagas' disease with evident cardiomyopathy, the kDNA minicircles integrate mainly into retrotransposons at several chromosomes, but the minicircles are also detected in coding regions of genes that regulate cell growth, differentiation, and immune responses. An accurate evaluation of the role played by the genotype alterations in the autoimmune rejection of self-tissues in Chagas' disease is achieved with the cross-kingdom chicken model system, which is refractory to T. cruzi infections. The inoculation of T. cruzi into embryonated eggs prior to incubation generates parasite-free chicks, which retain the kDNA minicircle sequence mainly in the macrochromosome coding genes. Crossbreeding transfers the kDNA mutations to the chicken progeny. The kDNA-mutated chickens develop severe cardiomyopathy in adult life and die of heart failure. The phenotyping of the lesions revealed that cytotoxic CD45, CD8(+) γδ, and CD8α(+) T lymphocytes carry out the rejection of the chicken heart. These results suggest that the inflammatory cardiomyopathy of Chagas' disease is a genetically driven autoimmune disease.
Collapse
Affiliation(s)
- Antonio R L Teixeira
- Chagas Disease Multidisciplinary Research Laboratory, University of Brasilia, Federal District, Brazil.
| | | | | | | | | |
Collapse
|
43
|
Pérez AR, Silva-Barbosa SD, Roggero E, Calmon-Hamaty F, Villar SR, Gutierrez FR, Silva JS, Savino W, Bottasso O. Immunoendocrinology of the thymus in Chagas disease. Neuroimmunomodulation 2011; 18:328-38. [PMID: 21952685 DOI: 10.1159/000329494] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
During immune response to infectious agents, the host develops an inflammatory response which could fail to eliminate the pathogen or may become dysregulated. In this case, the ongoing response acquires a new status and turns out to be detrimental. The same elements taking part in the establishment and regulation of the inflammatory response (cytokines, chemokines, regulatory T cells and counteracting compounds like glucocorticoids) may also mediate harmful effects. Thymic disturbances seen during Trypanosoma cruzi (T. cruzi) infection fit well with this conceptual framework. After infection, this organ suffers a severe atrophy due to apoptosis-induced thymocyte exhaustion, mainly affecting the immature double-positive (DP) CD4+CD8+ population. Thymus cellularity depletion, which occurs in the absence of main immunological mediators involved in anti-T. cruzi defense, seems to be linked to a systemic cytokine/hormonal imbalance, involving a dysregulated increase in Tumor Necrosis Factor alpha (TNF-α) and corticosterone hormone levels. Additionally, we have found an anomalous exit of potentially autoimmune DP cells to the periphery, in parallel to a shrinkage in the compartment of natural regulatory T cells. In this context, our data clearly point to the view that the thymus is a target organ of T. cruzi infection. Preserved thymus may be essential for the development of an effective immune response against T. cruzi, but this organ is severely affected by a dysregulated circuit of proinflammatory cytokines and glucocorticoids. Also, the alterations observed in the DP population might have potential implications for the autoimmune component of human Chagas disease.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Institute of Immunology, School of Medical Sciences, National University of Rosario (U.N.R.), Rosario, Argentina. perez_anarosa @ yahoo.com.ar
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Maioli TU, Assis FA, Vieira PMA, Borelli P, Santiago H, Alves R, Romanha AJ, Carneiro CM, Faria AMC. Splenectomy increases mortality in murine Trypanosoma cruzi infection. Scand J Immunol 2010; 73:36-45. [PMID: 21129001 DOI: 10.1111/j.1365-3083.2010.02478.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The spleen is a secondary lymphoid organ that harbours a variety of cells such as T and B lymphocytes and antigen-presenting cells important to immune response development. In this study, we evaluated the impact of spleen removal in the immune response to experimental Trypanosoma cruzi infection. C57BL/6 mice were infected with Y strain of the parasite and infection was followed daily. Mice that underwent splenectomy had fewer parasites in peripheral blood at the peak of infection; however, mortality was increased. Histological analysis of heart and liver tissues revealed an increased number of parasites and inflammatory infiltrates at these sites. Spleen removal was associated with reduction in IFN-γ and TNF-α production during infection as well as with a decrease in specific antibody secretion. Haematological disorders were also detected. Splenectomized mice exhibited severe anaemia and decreased bone marrow cell numbers. Our results indicate that spleen integrity is critical in T. cruzi infection for the immune response against the parasite, as well as for the control of bone marrow haematological function.
Collapse
Affiliation(s)
- T U Maioli
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nutritional imbalances and infections affect the thymus: consequences on T-cell-mediated immune responses. Proc Nutr Soc 2010; 69:636-43. [PMID: 20860857 DOI: 10.1017/s0029665110002545] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The thymus gland, where T lymphocyte development occurs, is targeted in malnutrition secondary to protein energy deficiency. There is a severe thymic atrophy, resulting from massive thymocyte apoptosis (particularly affecting the immature CD4+CD8+ cell subset) and decrease in cell proliferation. The thymic microenvironment (the non-lymphoid compartment that drives intrathymic T-cell development) is also affected in malnutrition: morphological changes in thymic epithelial cells were found, together with a decrease of thymic hormone production, as well as an increase of intrathymic contents of extracellular proteins. Profound changes in the thymus can also be seen in deficiencies of vitamins and trace elements. Taking Zn deficiency as an example, there is a substantial thymic atrophy. Importantly, marginal Zn deficiency in AIDS subjects, children with diarrhoea and elderly persons, significantly impairs the host's immunity, resulting in an increased risk of opportunistic infections and mortality; effects that are reversed by Zn supplementation. Thymic changes also occur in acute infectious diseases, including a severe thymic atrophy, mainly due to the depletion of CD4+CD8+ thymocytes, decrease in thymocyte proliferation, in parallel to densification of the epithelial network and increase in the extracellular matrix contents, with consequent disturbances in thymocyte migration and export. In conclusion, the thymus is targeted in several conditions of malnutrition as well as in acute infections. These changes are related to the impaired peripheral immune response seen in malnourished and infected individuals. Thus, strategies inducing thymus replenishment should be considered as adjuvant therapeutics to improve immunity in malnutrition and/or acute infectious diseases.
Collapse
|
46
|
Bermejo DA, Amezcua-Vesely MC, Montes CL, Merino MC, Gehrau RC, Cejas H, Acosta-Rodríguez EV, Gruppi A. BAFF mediates splenic B cell response and antibody production in experimental Chagas disease. PLoS Negl Trop Dis 2010; 4:e679. [PMID: 20454564 PMCID: PMC2864296 DOI: 10.1371/journal.pntd.0000679] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 03/24/2010] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND B cells and antibodies are involved not only in controlling the spread of blood circulating Trypanosoma cruzi, but also in the autoreactive manifestations observed in Chagas disease. Acute infection results in polyclonal B cell activation associated with hypergammaglobulinemia, delayed specific humoral immunity and high levels of non-parasite specific antibodies. Since TNF superfamily B lymphocyte Stimulator (BAFF) mediates polyclonal B cell response in vitro triggered by T. cruzi antigens, and BAFF-Tg mice show similar signs to T. cruzi infected mice, we hypothesized that BAFF can mediate polyclonal B cell response in experimental Chagas disease. METHODOLOGY/PRINCIPAL FINDINGS BAFF is produced early and persists throughout the infection. To analyze BAFF role in experimental Chagas disease, Balb/c infected mice were injected with BR3:Fc, a soluble receptor of BAFF, to block BAFF activity. By BAFF blockade we observed that this cytokine mediates the mature B cell response and the production of non-parasite specific IgM and IgG. BAFF also influences the development of antinuclear IgG and parasite-specific IgM response, not affecting T. cruzi-specific IgG and parasitemia. Interestingly, BAFF inhibition favors the parasitism in heart. CONCLUSIONS/SIGNIFICANCE Our results demonstrate, for the first time, an active role for BAFF in shaping the mature B cell repertoire in a parasite infection.
Collapse
Affiliation(s)
- Daniela A. Bermejo
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - María C. Amezcua-Vesely
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Carolina L. Montes
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - María C. Merino
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Ricardo C. Gehrau
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Hugo Cejas
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Eva V. Acosta-Rodríguez
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
| | - Adriana Gruppi
- Department of Immunology, School of Chemical Sciences, National University of Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|