1
|
Viano ME, Baez NS, Savid-Frontera C, Baigorri RE, Dinatale B, Pacini MF, Bulfoni Balbi C, Gonzalez FB, Fozzatti L, Lidón NL, Young HA, Hodge DL, Cerban F, Stempin CC, Pérez AR, Rodriguez-Galán MC. Systemic inflammatory Th1 cytokines during Trypanosoma cruzi infection disrupt the typical anatomical cell distribution and phenotypic/functional characteristics of various cell subsets within the thymus. Microbes Infect 2024; 26:105337. [PMID: 38615883 PMCID: PMC11227410 DOI: 10.1016/j.micinf.2024.105337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
The thymus plays a crucial role in T cell differentiation, a complex process influenced by various factors such as antigens, the microenvironment and thymic architecture. The way the thymus resolves infections is critical, as chronic persistence of microbes or inflammatory mediators can obstruct the differentiation. Here, we illustrate that following inflammatory T helper 1 infectious processes like those caused by Candida albicans or Trypanosoma cruzi, single positive thymocytes adopt a mature phenotype. Further investigations focused on T. cruzi infection, reveal a substantial existence of CD44+ cells in both the cortical and medullary areas of the thymus at the onset of infection. This disturbance coincides with heightened interferon gamma (IFNγ) production by thymocytes and an increased cytotoxic capacity against T. cruzi-infected macrophages. Additionally, we observe a reduced exportation capacity in T. cruzi-infected mice. Some alterations can be reversed in IFNγ knockout mice (KO). Notably, the majority of these effects can be replicated by systemic expression of interleukin (IL)-12+IL-18, underlining the predominantly inflammatory rather than pathogen-specific nature of these phenomena. Understanding the mechanisms through which systemic inflammation disrupts normal T cell development, as well as subsequent T cell exportation to secondary lymphoid organs (SLO) is pivotal for comprehending susceptibility to diseases in different pathological scenarios.
Collapse
Affiliation(s)
- Maria Estefania Viano
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Natalia Soledad Baez
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Constanza Savid-Frontera
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Ruth Eliana Baigorri
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Brenda Dinatale
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER CONICET-UNR), Argentina
| | - Maria Florencia Pacini
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER CONICET-UNR), Argentina
| | - Camila Bulfoni Balbi
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER CONICET-UNR), Argentina
| | | | - Laura Fozzatti
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Nicolas Leonel Lidón
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Howard A Young
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick MD 21702-1201, USA
| | - Deborah L Hodge
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick MD 21702-1201, USA
| | - Fabio Cerban
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Cinthia Carolina Stempin
- Inmunología, CIBICI-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Ana Rosa Pérez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER CONICET-UNR), Argentina; Centro de Investigación y Producción de Reactivos Biológicos (CIPREB), Facultad de Cs. Médicas de la Universidad Nacional de Rosario (UNR), Argentina
| | | |
Collapse
|
2
|
Lintao RCV, Kammala AK, Radnaa E, Bettayeb M, Vincent KL, Patrikeev I, Yaklic J, Bonney EA, Menon R. Characterization of fetal microchimeric immune cells in mouse maternal hearts during physiologic and pathologic pregnancies. Front Cell Dev Biol 2023; 11:1256945. [PMID: 37808080 PMCID: PMC10556483 DOI: 10.3389/fcell.2023.1256945] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction: During pregnancy, fetal cells can be incorporated into maternal tissues (fetal microchimerism), where they can persist postpartum. Whether these fetal cells are beneficial or detrimental to maternal health is unknown. This study aimed to characterize fetal microchimeric immune cells in the maternal heart during pregnancy and postpartum, and to identify differences in these fetal microchimeric subpopulations between normal and pregnancies complicated by spontaneous preterm induced by ascending infection. Methods: A Cre reporter mouse model, which when mated with wild-type C57BL/6J females resulted in cells and tissues of progeny expressing red fluorescent protein tandem dimer Tomato (mT+), was used to detect fetal microchimeric cells. On embryonic day (E)15, 104 colony-forming units (CFU) E. coli was administered intravaginally to mimic ascending infection, with delivery on or before E18.5 considered as preterm delivery. A subset of pregnant mice was sacrificed at E16 and postpartum day 28 to harvest maternal hearts. Heart tissues were processed for immunofluorescence microscopy and high-dimensional mass cytometry by time-of-flight (CyTOF) using an antibody panel of immune cell markers. Changes in cardiac physiologic parameters were measured up to 60 days postpartum via two-dimensional echocardiography. Results: Intravaginal E. coli administration resulted in preterm delivery of live pups in 70% of the cases. mT + expressing cells were detected in maternal uterus and heart, implying that fetal cells can migrate to different maternal compartments. During ascending infection, more fetal antigen-presenting cells (APCs) and less fetal hematopoietic stem cells (HSCs) and fetal double-positive (DP) thymocytes were observed in maternal hearts at E16 compared to normal pregnancy. These HSCs were cleared while DP thymocytes persisted 28 days postpartum following an ascending infection. No significant changes in cardiac physiologic parameters were observed postpartum except a trend in lowering the ejection fraction rate in preterm delivered mothers. Conclusion: Both normal pregnancy and ascending infection revealed distinct compositions of fetal microchimeric immune cells within the maternal heart, which could potentially influence the maternal cardiac microenvironment via (1) modulation of cardiac reverse modeling processes by fetal stem cells, and (2) differential responses to recognition of fetal APCs by maternal T cells.
Collapse
Affiliation(s)
- Ryan C. V. Lintao
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Enkhtuya Radnaa
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Mohamed Bettayeb
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Kathleen L. Vincent
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- Biomedical Engineering and Imaging Sciences Group, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Igor Patrikeev
- Biomedical Engineering and Imaging Sciences Group, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Jerome Yaklic
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Elizabeth A. Bonney
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Larner College of Medicine, The University of Vermont, Burlington, VT, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
3
|
Savino W, Lepletier A. Thymus-derived hormonal and cellular control of cancer. Front Endocrinol (Lausanne) 2023; 14:1168186. [PMID: 37529610 PMCID: PMC10389273 DOI: 10.3389/fendo.2023.1168186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
The thymus gland is a central lymphoid organ in which developing T cell precursors, known as thymocytes, undergo differentiation into distinct type of mature T cells, ultimately migrating to the periphery where they exert specialized effector functions and orchestrate the immune responses against tumor cells, pathogens and self-antigens. The mechanisms supporting intrathymic T cell differentiation are pleiotropically regulated by thymic peptide hormones and cytokines produced by stromal cells in the thymic microenvironment and developing thymocytes. Interestingly, in the same way as T cells, thymic hormones (herein exemplified by thymosin, thymulin and thymopoietin), can circulate to impact immune cells and other cellular components in the periphery. Evidence on how thymic function influences tumor cell biology and response of patients with cancer to therapies remains unsatisfactory, although there has been some improvement in the knowledge provided by recent studies. Herein, we summarize research progression in the field of thymus-mediated immunoendocrine control of cancer, providing insights into how manipulation of the thymic microenvironment can influence treatment outcomes, including clinical responses and adverse effects of therapies. We review data obtained from clinical and preclinical cancer research to evidence the complexity of immunoendocrine interactions underpinning anti-tumor immunity.
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
- INOVA-IOC Network on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ailin Lepletier
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
4
|
Puerta CJ, Cuellar A, Lasso P, Mateus J, Gonzalez JM. Trypanosoma cruzi-specific CD8 + T cells and other immunological hallmarks in chronic Chagas cardiomyopathy: Two decades of research. Front Cell Infect Microbiol 2023; 12:1075717. [PMID: 36683674 PMCID: PMC9846209 DOI: 10.3389/fcimb.2022.1075717] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023] Open
Abstract
Trypanosoma cruzi, the causal agent of Chagas disease, has coexisted with humans for thousands of years. Therefore, the parasite has developed several mechanisms of antigenic variability that has allowed it to live inside the cells and evade the host immune response. Since T. cruzi displays an intracellular cycle-stage, our research team focused on providing insights into the CD8+ T cells immune response in chronic Chagas cardiomyopathy. We began our work in the 2000s studying parasite antigens that induce natural immune responses such as the KMP11 protein and TcTLE, its N-terminal derived peptide. Different approaches allowed us to reveal TcTLE peptide as a promiscuous CD8+ T cell epitope, able of inducing multifunctional cellular immune responses and eliciting a humoral response capable of decreasing parasite movement and infective capacity. Next, we demonstrated that as the disease progresses, total CD8+ T cells display a dysfunctional state characterized by a prolonged hyper-activation state along with an increase of inhibitory receptors (2B4, CD160, PD-1, TIM-3, CTLA-4) expression, an increase of specific terminal effector T cells (TTE), a decrease of proliferative capacity, a decrease of stem cell memory (TSCM) frequency, and a decrease of CD28 and CD3ζ expression. Thus, parasite-specific CD8+ T cells undergo clonal exhaustion, distinguished by an increase in late-differentiated cells, a mono-functional response, and enhanced expression of inhibitory receptors. Finally, it was found that anti-parasitic treatment induces an improved CD8+ T cell response in asymptomatic individuals, and a mouse animal model led us to establish a correlation between the quality of the CD8+ T cell responses and the outcome of chronic infection. In the future, using OMICs strategies, the identification of the specific cellular signals involved in disease progression will provide an invaluable resource for discovering new biomarkers of progression or new vaccine and immunotherapy strategies. Also, the inclusion of the TcTLE peptide in the rational design of epitope-based vaccines, the development of immunotherapy strategies using TSCM or the blocking of inhibitory receptors, and the use of the CD8+ T cell response quality to follow treatments, immunotherapies or vaccines, all are alternatives than could be explored in the fight against Chagas disease.
Collapse
Affiliation(s)
- Concepción J. Puerta
- Laboratory of Molecular Parasitology, Infectious Diseases Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Adriana Cuellar
- Clinical Laboratory Sciences Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Paola Lasso
- Laboratory of Molecular Parasitology, Infectious Diseases Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Jose Mateus
- Laboratory of Molecular Parasitology, Infectious Diseases Group, Department of Microbiology, School of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - John M. Gonzalez
- Group of Biomedical Sciences, School of Medicine, Universidad de Los Andes, Bogotá, Colombia
| |
Collapse
|
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
|
Mileto SJ, Hutton ML, Walton SL, Das A, Ioannidis LJ, Ketagoda D, Quinn KM, Denton KM, Hansen DS, Lyras D. Bezlotoxumab prevents extraintestinal organ damage induced by Clostridioides difficile infection. Gut Microbes 2022; 14:2117504. [PMID: 36045589 PMCID: PMC9450906 DOI: 10.1080/19490976.2022.2117504] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Clostridioides difficile is the most common cause of infectious antibiotic-associated diarrhea, with disease mediated by two major toxins TcdA and TcdB. In severe cases, systemic disease complications may arise, resulting in fatal disease. Systemic disease in animal models has been described, with thymic damage an observable consequence of severe disease in mice. Using a mouse model of C. difficile infection, we examined this disease phenotype, focussing on the thymus and serum markers of systemic disease. The efficacy of bezlotoxumab, a monoclonal TcdB therapeutic, to prevent toxin mediated systemic disease complications was also examined. C. difficile infection causes toxin-dependent thymic damage and CD4+CD8+ thymocyte depletion in mice. These systemic complications coincide with changes in biochemical markers of liver and kidney function, including increased serum urea and creatinine, and hypoglycemia. Administration of bezlotoxumab during C. difficile infection prevents systemic disease and thymic atrophy, without blocking gut damage, suggesting the leakage of gut contents into circulation may influence systemic disease. As the thymus has such a crucial role in T cell production and immune system development, these findings may have important implications in relapse of C. difficile disease and impaired immunity during C. difficile infection. The prevention of thymic atrophy and reduced systemic response following bezlotoxumab treatment, without altering colonic damage, highlights the importance of systemic disease in C. difficile infection, and provides new insights into the mechanism of action for this therapeutic.Abbreviations: Acute kidney injury (AKI); Alanine Transaminase (ALT); Aspartate Aminotransferase (AST); C. difficile infection (CDI); chronic kidney disease (CKD); combined repetitive oligo-peptides (CROPS); cardiovascular disease (CVD); Double positive (DP); hematoxylin and eosin (H&E); immunohistochemical (IHC); multiple organ dysfunction syndrome (MODS); phosphate buffered saline (PBS); standard error of the mean (SEM); surface layer proteins (SLP); Single positive (SP); wild-type (WT).
Collapse
Affiliation(s)
- Steven J. Mileto
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Melanie L. Hutton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Sarah L. Walton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Antariksh Das
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Lisa J. Ioannidis
- Walter and Eliza Hall Insitiute, Infectious Diseases and Immune Defence Division, Parkville, Australia,Department of Medical Biology, the University of Melbourne, Parkville, Australia
| | - Don Ketagoda
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia
| | - Kylie M. Quinn
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia,RMIT University School of Biomedical and Health Sciences, Chronic Inflammatory and Infectious Diseases Program, Bundoora, Australia
| | - Kate M. Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Australia
| | - Diana S. Hansen
- Walter and Eliza Hall Insitiute, Infectious Diseases and Immune Defence Division, Parkville, Australia,Department of Medical Biology, the University of Melbourne, Parkville, Australia
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Australia,CONTACT Dena Lyras Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria3800, Australia
| |
Collapse
|
7
|
Barreira-Silva P, Melo-Miranda R, Nobrega C, Roque S, Serre-Miranda C, Borges M, Armada G, de Sá Calçada D, Behar SM, Appelberg R, Correia-Neves M. IFNγ and iNOS-Mediated Alterations in the Bone Marrow and Thymus and Its Impact on Mycobacterium avium-Induced Thymic Atrophy. Front Immunol 2021; 12:696415. [PMID: 34987496 PMCID: PMC8721011 DOI: 10.3389/fimmu.2021.696415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Disseminated infection with the high virulence strain of Mycobacterium avium 25291 leads to progressive thymic atrophy. We previously showed that M. avium-induced thymic atrophy results from increased glucocorticoid levels that synergize with nitric oxide (NO) produced by interferon gamma (IFNγ) activated macrophages. Where and how these mediators act is not understood. We hypothesized that IFNγ and NO promote thymic atrophy through their effects on bone marrow (BM) T cell precursors and T cell differentiation in the thymus. We show that M. avium infection cause a reduction in the percentage and number of common lymphoid progenitors (CLP). Additionally, BM precursors from infected mice show an overall impaired ability to reconstitute thymi of RAGKO mice, in part due to IFNγ. Thymi from infected mice present an IFNγ and NO-driven inflammation. When transplanted under the kidney capsule of uninfected mice, thymi from infected mice are unable to sustain T cell differentiation. Finally, we observed increased thymocyte death via apoptosis after infection, independent of both IFNγ and iNOS; and a decrease on active caspase-3 positive thymocytes, which is not observed in the absence of iNOS expression. Together our data suggests that M. avium-induced thymic atrophy results from a combination of defects mediated by IFNγ and NO, including alterations in the BM T cell precursors, the thymic structure and the thymocyte differentiation.
Collapse
Affiliation(s)
- Palmira Barreira-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
- *Correspondence: Palmira Barreira-Silva, ; Margarida Correia-Neves,
| | - Rita Melo-Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Claudia Nobrega
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Susana Roque
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Cláudia Serre-Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Margarida Borges
- Research Unit on Applied Molecular Biosciences (UCIBIO)/Rede de Química e Tecnologia (REQUINTE), Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Gisela Armada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Daniela de Sá Calçada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
| | - Samuel M. Behar
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Rui Appelberg
- Instituto de Investigação e Inovação em Saúde (i3S), Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics Research Group (ICVS/3B’s), PT Government Associate Laboratory, Braga, Portugal
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Palmira Barreira-Silva, ; Margarida Correia-Neves,
| |
Collapse
|
8
|
Garcia-Bustos V, Moral Moral P, Cabañero-Navalon MD, Salavert Lletí M, Calabuig Muñoz E. Does Autoimmunity Play a Role in the Immunopathogenesis of Vasculitis Associated With Chronic Chagas Disease? Front Cell Infect Microbiol 2021; 11:671962. [PMID: 34295833 PMCID: PMC8290184 DOI: 10.3389/fcimb.2021.671962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 06/18/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Victor Garcia-Bustos
- Department of Internal Medicine and Infectious Diseases, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Pedro Moral Moral
- Department of Internal Medicine and Infectious Diseases, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Marta Dafne Cabañero-Navalon
- Department of Internal Medicine and Infectious Diseases, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Miguel Salavert Lletí
- Department of Internal Medicine and Infectious Diseases, University and Polytechnic La Fe Hospital, Valencia, Spain
| | - Eva Calabuig Muñoz
- Department of Internal Medicine and Infectious Diseases, University and Polytechnic La Fe Hospital, Valencia, Spain.,Department of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
9
|
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
|
10
|
Nájera CA, Batista MF, Meneghelli I, Bahia D. Mixed signals - how Trypanosoma cruzi exploits host-cell communication and signaling to establish infection. J Cell Sci 2021; 134:134/5/jcs255687. [PMID: 33692153 DOI: 10.1242/jcs.255687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chagas disease (American trypanosomiasis) is a 'neglected' pathology that affects millions of people worldwide, mainly in Latin America. Trypanosoma cruzi, the causative agent, is an obligate intracellular parasite with a complex and diverse biology that infects several mammalian species, including humans. Because of genetic variability among strains and the presence of four biochemically and morphologically distinct parasite forms, the outcome of T. cruzi infection varies considerably depending on host cell type and parasite strain. During the initial contact, cellular communication is established by host-recognition-mediated responses, followed by parasite adherence and penetration. For this purpose, T. cruzi expresses a variety of proteins that modify the host cell, enabling it to safely reach the cytoplasm. After entry into the host cell, T. cruzi forms a transitory structure termed 'parasitophorous vacuole' (PV), followed by its cytoplasmic replication and differentiation after PV rupture, and subsequent invasion of other cells. The success of infection, maintenance and survival inside host cells is facilitated by the ability of T. cruzi to subvert various host signaling mechanisms. We focus in this Review on the various mechanisms that induce host cytoskeletal rearrangements, activation of autophagy-related proteins and crosstalk among major immune response regulators, as well as recent studies on the JAK-STAT pathway.
Collapse
Affiliation(s)
- Carlos Acides Nájera
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Marina Ferreira Batista
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Isabela Meneghelli
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| | - Diana Bahia
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 486, Brazil
| |
Collapse
|
11
|
Savino W, Chaves B, Bonomo AC, Cotta-de-Almeida V. Integrin-directed antibody-based immunotherapy: focus on VLA-4. IMMUNOTHERAPY ADVANCES 2021; 1:ltab002. [PMID: 35919739 PMCID: PMC9327104 DOI: 10.1093/immadv/ltab002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/13/2020] [Accepted: 01/13/2021] [Indexed: 12/28/2022] Open
Abstract
One major finding of chronic inflammatory diseases of various origins is the establishment of inflammatory infiltrates, bearing different leukocyte subpopulations, including activated T lymphocytes. Integrins are among the large series of molecular interactions that have been implicated as players in both triggering and maintenance of leukocyte influx from the blood into a given organ parenchyme. Accordingly, blocking the interaction between VLA-6 integrin and laminin, experimentally abrogates heart graft rejection. Many reports have shown that VLA-4 is used by T cells to cross endothelial barriers, as well as to migrate within target tissues. In this respect, a humanized IgG4 anti-VLA-4 monoclonal antibody (specific to the α4-integrin chain of VLA-4) has been successfully applied to treat multiple sclerosis as well as inflammatory bowel disease. Anti-VLA-4 monoclonal antibody has also been applied to block transendothelial passage in other autoimmune diseases, such as rheumatoid arthritis. On this same vein is the action of such a reagent in impairing in vitro transendothial and fibronectin-driven migration of CD4+ and CD8+ T cells expressing high densities of VLA-4 from Duchenne muscular dystrophy patients, thus potentially enlarging the use of this strategy to other diseases. Yet, in a small number of patients, the use of Natalizumab has been correlated with the progressive multifocal leukoencephalopathy, a serious brain infection caused by the John Cunningham virus. This issue restricted the use of the reagent. In this respect, the development of smaller and more specific antibody reagents should be envisioned as a next-generation promising strategy.
Collapse
Affiliation(s)
- 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), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Beatriz Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Computational Modeling Group, Oswaldo Cruz Foundation, Eusébio, Ceará, Brazil
| | - Adriana Cesar Bonomo
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vinicius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Brazão V, Colato RP, Santello FH, Vale GTD, Gonzaga NDA, Tirapelli CR, Prado JCD. Effects of melatonin on thymic and oxidative stress dysfunctions during Trypanosoma cruzi infection. J Pineal Res 2018; 65:e12510. [PMID: 29781553 DOI: 10.1111/jpi.12510] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/09/2018] [Indexed: 12/11/2022]
Abstract
Although the exact etiology of Chagas disease is not completely elucidated, thymic atrophy and oxidative stress are believed to be important contributors to the pathogenesis during acute Trypanosoma cruzi (T. cruzi) infection. We hypothesized that exogenous melatonin, administered by gavage (5 mg/kg, p.o., gavage) to young (5 weeks old) and middle-aged (18 months old) male Wistar rats, would modulate thymic oxidative damage and reverse the age-related thymus regression during T. cruzi acute infection. Increased levels of superoxide anion (O2- ) were detected in the thymus of infected animals, and treatment with melatonin reverted this response. We found reduced TBARS levels as well as a significant increase in superoxide dismutase (SOD) activity in the thymus of all middle-aged melatonin-treated animals, infected or not with T. cruzi. Furthermore, melatonin increased the thymic expression of SOD1 and SOD2 in middle-aged control animals. Nox2 expression was not affected by melatonin treatment in young or middle-aged animals. Melatonin reverted the age-related thymic regression as revealed by the increase in thymus weight, total number of thymocytes, and reduction in age-related accumulation of double-negative thymocytes. This is the first report to directly examine the effects of melatonin treatment on the thymic antioxidant/oxidant status and thymic changes during T. cruzi infection. Our results revealed new antioxidant features that turn melatonin a potentially useful compound for the treatment of Chagas disease, a condition in which an excessive oxidative damage occurs.
Collapse
Affiliation(s)
- Vânia Brazão
- College of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela Pravato Colato
- College of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Fabricia Helena Santello
- College of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Gabriel Tavares do Vale
- Department of Psychiatric Nursing and Human Sciences, Laboratory of Pharmacology, College of Nursing of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Natália de Almeida Gonzaga
- Department of Psychiatric Nursing and Human Sciences, Laboratory of Pharmacology, College of Nursing of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Carlos Renato Tirapelli
- Department of Psychiatric Nursing and Human Sciences, Laboratory of Pharmacology, College of Nursing of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - José Clóvis do Prado
- College of Pharmaceutical Sciences of Ribeirão Preto (FCFRP), University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
15
|
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
|
16
|
Leptin regulates Granzyme-A, PD-1 and CTLA-4 expression in T cell to control visceral leishmaniasis in BALB/c Mice. Sci Rep 2017; 7:14664. [PMID: 29116252 PMCID: PMC5676676 DOI: 10.1038/s41598-017-15288-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/24/2017] [Indexed: 01/01/2023] Open
Abstract
Visceral leishmaniasis (VL) is responsible for several deaths in malnourished children accompanied by diminished circulating leptin and impaired cell-mediated immunity. Typically, leptin deficiency is associated with the Th2 polarization that markedly coincides with the pathogenesis of VL. The aim of the present study was to unravel the prophylactic role of leptin in malnutrition-coupled VL mice. Interestingly, we observed that L. donovani infection itself reduces the serum leptin levels in malnutrition. Exogenous leptin restored severe body weight loss and parasite load in the spleen and liver of malnourished infected mice compared to controls. Leptin increases functional CD8+ T-cell population, Granzyme-A expression down-regulates anergic T-cell markers such as PD-1 and CTLA-4. It was also noticed that, leptin suppresses GM-CSF mRNA expression in parasite favored monocytes and reduced arginase activity in bone marrow derived macrophage indicate macrophages dependent T-cell activation and proliferation. Leptin-induced IFN-γ, IL-2, and TNF-α cytokines in the culture supernatant of splenocytes upon soluble leishmanial antigen (SLA) stimulation and significantly up-regulates serum IgG2a titers, which help to generate Th1 immune response in VL. Furthermore, leptin induced a granulomatous response and restored L. donovani induced tissue degeneration in the liver. Altogether, our findings suggest the exogenous leptin can restore T cell mediated immunity in malnourished VL mice.
Collapse
|
17
|
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
|
18
|
Savino W, Mendes-da-Cruz DA, Lepletier A, Dardenne M. Hormonal control of T-cell development in health and disease. Nat Rev Endocrinol 2016; 12:77-89. [PMID: 26437623 DOI: 10.1038/nrendo.2015.168] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The physiology of the thymus, the primary lymphoid organ in which T cells are generated, is controlled by hormones. Data from animal models indicate that several peptide and nonpeptide hormones act pleiotropically within the thymus to modulate the proliferation, differentiation, migration and death by apoptosis of developing thymocytes. For example, growth hormone and prolactin can enhance thymocyte proliferation and migration, whereas glucocorticoids lead to the apoptosis of these developing cells. The thymus undergoes progressive age-dependent atrophy with a loss of cells being generated and exported, therefore, hormone-based therapies are being developed as an alternative strategy to rejuvenate the organ, as well as to augment thymocyte proliferation and the export of mature T cells to peripheral lymphoid organs. Some hormones (such as growth hormone and progonadoliberin-1) are also being used as therapeutic agents to treat immunodeficiency disorders associated with thymic atrophy, such as HIV infection. In this Review, we discuss the accumulating data that shows the thymus gland is under complex and multifaceted hormonal control that affects the process of T-cell development in health and disease.
Collapse
Affiliation(s)
- Wilson Savino
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenue Brasil 4365, 21045-900, Manguinhos, Rio de Janeiro, Brazil
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenue Brasil 4365, 21045-900, Manguinhos, Rio de Janeiro, Brazil
| | - Ailin Lepletier
- Laboratory of Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenue Brasil 4365, 21045-900, Manguinhos, Rio de Janeiro, Brazil
| | - Mireille Dardenne
- Hôpital Necker, CNRS UMR 8147, Université Paris Descartes, 75015 Paris, France
| |
Collapse
|
19
|
González FB, Calmon-Hamaty F, Nô Seara Cordeiro S, Fernández Bussy R, Spinelli SV, D'Attilio L, Bottasso O, Savino W, Cotta-de-Almeida V, Villar SR, Pérez AR. Trypanosoma cruzi Experimental Infection Impacts on the Thymic Regulatory T Cell Compartment. PLoS Negl Trop Dis 2016; 10:e0004285. [PMID: 26745276 PMCID: PMC4706328 DOI: 10.1371/journal.pntd.0004285] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/16/2015] [Indexed: 11/18/2022] Open
Abstract
The dynamics of regulatory T cells in the course of Trypanosoma cruzi infection is still debated. We previously demonstrated that acute murine T. cruzi infection results in an impaired peripheral CD4+Foxp3+ T cell differentiation due to the acquisition of an abnormal Th1-like phenotype and altered functional features, negatively impacting on the course of infection. Moreover, T. cruzi infection induces an intense thymic atrophy. As known, the thymus is the primary lymphoid organ in which thymic-derived regulatory T cells, known as tTregs, differentiate. Considering the lack of available data about the effect of T. cruzi infection upon tTregs, we examined tTreg dynamics during the course of disease. We confirmed that T. cruzi infection induces a marked loss of tTreg cell number associated to cell precursor exhaustion, partially avoided by glucocorticoid ablation- and IL-2 survival factor depletion. At the same time, tTregs accumulate within the CD4 single-positive compartment, exhibiting an increased Ki-67/Annexin V ratio compared to controls. Moreover, tTregs enhance after the infection the expression of signature markers (CD25, CD62L and GITR) and they also display alterations in the expression of migration-associated molecules (α chains of VLAs and chemokine receptors) such as functional fibronectin-driven migratory disturbance. Taken together, we provide data demonstrating profound alterations in tTreg compartment during acute murine T. cruzi infection, denoting that their homeostasis is significantly affected. The evident loss of tTreg cell number may compromise the composition of tTreg peripheral pool, and such sustained alteration over time may be partially related to the immune dysregulation observed in the chronic phase of the disease.
Collapse
Affiliation(s)
- Florencia Belén González
- Institute of Clinical and Experimental Immunology of Rosario (IDICER CONICET-UNR), Rosario, Argentina
| | - Flavia Calmon-Hamaty
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Synara Nô Seara Cordeiro
- Laboratory of Innovations in Therapy, Teaching and Bioproducts, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Rodrigo Fernández Bussy
- Institute of Clinical and Experimental Immunology of Rosario (IDICER CONICET-UNR), Rosario, Argentina
| | - Silvana Virginia Spinelli
- Institute of Clinical and Experimental Immunology of Rosario (IDICER CONICET-UNR), Rosario, Argentina
| | - Luciano D'Attilio
- Institute of Clinical and Experimental Immunology of Rosario (IDICER CONICET-UNR), Rosario, Argentina
| | - Oscar Bottasso
- Institute of Clinical and Experimental Immunology of Rosario (IDICER CONICET-UNR), Rosario, Argentina
| | - 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
| | - Silvina Raquel Villar
- Institute of Clinical and Experimental Immunology of Rosario (IDICER CONICET-UNR), Rosario, Argentina
| | - Ana Rosa Pérez
- Institute of Clinical and Experimental Immunology of Rosario (IDICER CONICET-UNR), Rosario, Argentina
- * E-mail: ,
| |
Collapse
|
20
|
Savino W, Mendes-da-Cruz DA, Golbert DCF, Riederer I, Cotta-de-Almeida V. Laminin-Mediated Interactions in Thymocyte Migration and Development. Front Immunol 2015; 6:579. [PMID: 26635793 PMCID: PMC4648024 DOI: 10.3389/fimmu.2015.00579] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/29/2015] [Indexed: 11/30/2022] Open
Abstract
Intrathymic T-cell differentiation is a key process for the development and maintenance of cell-mediated immunity, and occurs concomitantly to highly regulated migratory events. We have proposed a multivectorial model for describing intrathymic thymocyte migration. One of the individual vectors comprises interactions mediated by laminins (LMs), a heterotrimeric protein family of the extracellular matrix. Several LMs are expressed in the thymus, being produced by microenvironmental cells, particularly thymic epithelial cells (TECs). Also, thymocytes and epithelial cells express integrin-type LM receptors. Functionally, it has been reported that the dy/dy mutant mouse (lacking the LM isoform 211) exhibits defective thymocyte differentiation. Several data show haptotactic effects of LMs upon thymocytes, as well as their adhesion on TECs; both effects being prevented by anti-LM or anti-LM receptor antibodies. Interestingly, LM synergizes with chemokines to enhance thymocyte migration, whereas classe-3 semaphorins and B ephrins, which exhibit chemorepulsive effects in the thymus, downregulate LM-mediated migratory responses of thymocytes. More recently, we showed that knocking down the ITGA6 gene (which encodes the α6 integrin chain of LM receptors) in human TECs modulates a large number of cell migration-related genes and results in changes of adhesion pattern of thymocytes onto the thymic epithelium. Overall, LM-mediated interactions can be placed at the cross-road of the multivectorial process of thymocyte migration, with a direct influence per se, as well as by modulating other molecular interactions associated with the intrathymic-trafficking events.
Collapse
Affiliation(s)
- Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | | | | | - Ingo Riederer
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | - Vinicius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| |
Collapse
|
21
|
Linhares-Lacerda L, Palu CC, Ribeiro-Alves M, Paredes BD, Morrot A, Garcia-Silva MR, Cayota A, Savino W. Differential Expression of microRNAs in Thymic Epithelial Cells from Trypanosoma cruzi Acutely Infected Mice: Putative Role in Thymic Atrophy. Front Immunol 2015; 6:428. [PMID: 26347748 PMCID: PMC4543887 DOI: 10.3389/fimmu.2015.00428] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/06/2015] [Indexed: 12/21/2022] Open
Abstract
A common feature seen in acute infections is a severe atrophy of the thymus. This occurs in the murine model of acute Chagas disease. Moreover, in thymuses from Trypanosoma cruzi acutely infected mice, thymocytes exhibit an increase in the density of fibronectin and laminin integrin-type receptors, with an increase in migratory response ex vivo. Thymic epithelial cells (TEC) play a major role in the intrathymic T cell differentiation. To date, the consequences of molecular changes promoted by parasite infection upon thymus have not been elucidated. Considering the importance of microRNA for gene expression regulation, 85 microRNAs (mRNAs) were analyzed in TEC from T. cruzi acutely infected mice. The infection significantly modulated 29 miRNAs and modulation of 9 was also dependent whether TEC sorted out from the thymus exhibited cortical or medullary phenotype. In silico analysis revealed that these miRNAs may control target mRNAs known to be responsible for chemotaxis, cell adhesion, and cell death. Considering that we sorted TEC in the initial phase of thymocyte loss, it is conceivable that changes in TEC miRNA expression profile are functionally related to thymic atrophy, providing new clues to better understanding the mechanisms of the thymic involution seen in experimental Chagas disease.
Collapse
Affiliation(s)
- Leandra Linhares-Lacerda
- Laboratory on Thymus Research, Institute Oswaldo Cruz, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | - Cintia Cristina Palu
- Laboratory on Thymus Research, Institute Oswaldo Cruz, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | - Marcelo Ribeiro-Alves
- HIV/AIDS Clinical Research Center, National Institute of Infectious Diseases, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | - Bruno Diaz Paredes
- The National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Alexandre Morrot
- Department of Immunology, Microbiology Institute, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | | | - Alfonso Cayota
- Functional Genomics Unit, Institut Pasteur de Montevideo , Montevideo , Uruguay
| | - Wilson Savino
- Laboratory on Thymus Research, Institute Oswaldo Cruz, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| |
Collapse
|
22
|
Modulation of Intrathymic Sphingosine-1-Phosphate Levels Promotes Escape of Immature Thymocytes to the Periphery with a Potential Proinflammatory Role in Chagas Disease. BIOMED RESEARCH INTERNATIONAL 2015; 2015:709846. [PMID: 26347020 PMCID: PMC4539443 DOI: 10.1155/2015/709846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/21/2015] [Indexed: 11/30/2022]
Abstract
The sphingosine-1-phosphate (S1P) system regulates both thymic and lymph nodes T cell egress which is essential for producing and maintaining the recycling T cell repertoire. Infection with the protozoan parasite Trypanosoma cruzi induces a hormonal systemic deregulation that has impact in the thymic S1P homeostasis that ultimately promotes the premature exit of immature CD4−CD8− T cells expressing TCR and proinflamatory cytokines to peripheral lymphoid organs, where they may interfere with adaptive immune responses. In what follows, we review recent findings revealing escape of these immature T cells exhibiting an activation profile to peripheral compartments of the immune system in both experimental murine and human models of Chagas disease.
Collapse
|
23
|
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
|
24
|
Farias-de-Oliveira DA, Cotta-de-Almeida V, Villa-Verde DMS, Riederer I, Meis JD, Savino W. Fibronectin modulates thymocyte-thymic epithelial cell interactions following Trypanosoma cruzi infection. Mem Inst Oswaldo Cruz 2015; 108:825-31. [PMID: 24271041 PMCID: PMC3970635 DOI: 10.1590/0074-0276130071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 08/16/2013] [Indexed: 01/15/2023] Open
Abstract
Developing thymocytes interact with thymic epithelial cells (TECs) through cell-cell
interactions, TEC-derived secretory moieties and extracellular matrix (ECM)-mediated
interactions. These physiological interactions are crucial for normal thymocyte
differentiation, but can be disrupted in pathological situations. Indeed, there is severe
thymic atrophy in animals acutely infected with Trypanosoma cruzi due to
CD4+CD8+ thymocyte depletion secondary to caspase-mediated apoptosis, together with
changes in ECM deposition and thymocyte migration. We studied an in vitro model of TEC
infection by T. cruzi and found that infected TEC cultures show a reduced
number of cells, which was likely associated with decreased proliferative capacity, but
not with increased cell death, as demonstrated by bromodeoxyuridine and annexin-V
labelling. The infected TEC cultures exhibited increased expression of fibronectin (FN),
laminin (LM) and type IV collagen. Importantly, treatment with FN increased the relative
number of infected cells, whereas treatment with anti-FN or anti-LM antibodies resulted in
lower infection rates. Consistent with these data, we observed increased thymocyte
adhesion to infected TEC cultures. Overall, these results suggest that ECM molecules,
particularly FN, facilitate infection of the thymic epithelium and that the consequent
enhancement of ECM expression might be associated with changes in TEC-thymocyte
interactions.
Collapse
|
25
|
Cuervo-Escobar S, Losada-Barragán M, Umaña-Pérez A, Porrozzi R, Saboia-Vahia L, Miranda LHM, Morgado FN, Menezes RC, Sánchez-Gómez M, Cuervo P. T-cell populations and cytokine expression are impaired in thymus and spleen of protein malnourished BALB/c mice infected with Leishmania infantum. PLoS One 2014; 9:e114584. [PMID: 25535967 PMCID: PMC4275170 DOI: 10.1371/journal.pone.0114584] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/11/2014] [Indexed: 01/04/2023] Open
Abstract
Visceral leishmaniasis (VL) is a parasitic infectious disease that causes significant morbidity and mortality in the tropical and subtropical regions of the world. Although infections with visceralizing Leishmania may be asymptomatic, factors such as undernutrition increase the likelihood of progressing to clinical disease. Protein malnutrition, the most deleterious cause of malnutrition in developing countries, has been considered as a primary risk factor for the development of clinical VL. However, data regarding the immunological basis of this association are scarce. With the aim to analyze the effects of protein malnutrition on Leishmania infantum infection, we used BALB/c mice subjected to control or low protein isocaloric diets. Each animal group was divided into two subgroups and one was infected with L. infantum resulting in four study groups: animals fed 14% protein diet (CP), animals fed 4% protein diet (LP), animals fed 14% protein diet and infected (CPi), and animals fed 4% protein diet and infected (LPi).The susceptibility to L. infantum infection and immune responses were assessed in terms of body and lymphoid organ weight, parasite load, lymphocyte subpopulations, and cytokine expression. LPi mice had a significant reduction of body and lymphoid organ weight and exhibited a severe decrease of lymphoid follicles in the spleen. Moreover, LPi animals showed a significant decrease in CD4+CD8+ T cells in the thymus, whereas there was an increase of CD4+ and CD8+ T cells percentages in the spleen. Notably, the cytokine mRNA levels in the thymus and spleen of protein malnourished-infected animals were altered compared to the CP mice. Protein malnutrition results in a drastic dysregulation of T cells and cytokine expression in the thymus and spleen of L. infantum-infected BALB/c mice, which may lead to defective regulation of the thymocyte population and an impaired splenic immune response, accelerating the events of a normal course of infection.
Collapse
Affiliation(s)
- Sergio Cuervo-Escobar
- Laboratorio de Hormonas, Departamento de Química, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Monica Losada-Barragán
- Laboratorio de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Adriana Umaña-Pérez
- Laboratorio de Hormonas, Departamento de Química, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Renato Porrozzi
- Laboratorio de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Leonardo Saboia-Vahia
- Laboratorio de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Luisa H M Miranda
- Instituto de Pesquisa Clínica Evandro Chagas, IPEC, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Fernanda N Morgado
- Laboratorio de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Rodrigo C Menezes
- Instituto de Pesquisa Clínica Evandro Chagas, IPEC, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Myriam Sánchez-Gómez
- Laboratorio de Hormonas, Departamento de Química, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Patricia Cuervo
- Laboratorio de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
26
|
Lepletier A, de Almeida L, Santos L, da Silva Sampaio L, Paredes B, González FB, Freire-de-Lima CG, Beloscar J, Bottasso O, Einicker-Lamas M, Pérez AR, Savino W, Morrot A. Early double-negative thymocyte export in Trypanosoma cruzi infection is restricted by sphingosine receptors and associated with human chagas disease. PLoS Negl Trop Dis 2014; 8:e3203. [PMID: 25330249 PMCID: PMC4199546 DOI: 10.1371/journal.pntd.0003203] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 08/20/2014] [Indexed: 12/20/2022] Open
Abstract
The protozoan parasite Trypanosoma cruzi is able to target the thymus and induce alterations of the thymic microenvironmental and lymphoid compartments. Acute infection results in severe atrophy of the organ and early release of immature thymocytes into the periphery. To date, the pathophysiological effects of thymic changes promoted by parasite-inducing premature release of thymocytes to the periphery has remained elusive. Herein, we show that sphingosine-1-phosphate (S1P), a potent mediator of T cell chemotaxis, plays a role in the exit of immature double-negative thymocytes in experimental Chagas disease. In thymuses from T. cruzi-infected mice we detected reduced transcription of the S1P kinase 1 and 2 genes related to S1P biosynthesis, together with increased transcription of the SGPL1 sphingosine-1-lyase gene, whose product inactivates S1P. These changes were associated with reduced intrathymic levels of S1P kinase activity. Interestingly, double-negative thymocytes from infected animals expressed high levels of the S1P receptor during infection, and migrated to lower levels of S1P. Moreover, during T. cruzi infection, this thymocyte subset expresses high levels of IL-17 and TNF-α cytokines upon polyclonal stimulation. In vivo treatment with the S1P receptor antagonist FTY720 resulted in recovery the numbers of double-negative thymocytes in infected thymuses to physiological levels. Finally, we showed increased numbers of double-negative T cells in the peripheral blood in severe cardiac forms of human Chagas disease. The formation of mature lineage-committed T cells requires the specialized environment of the thymus, a central organ of the immune system supporting the development of self-tolerant T cells. Key events of intrathymic T-cell development include lineage commitment, selection events and thymic emigration. This organ undergoes physiological involution during aging. However, acute thymic atrophy can occur in the presence autoimmune diseases, malignant tumors and infections caused by intracellular pathogens. The present study shows that the protozoan parasite Trypanosoma cruzi changes the thymic microenvironmental and lymphoid compartments, resulting in premature release of very immature CD4−CD8− double-negative thymocytes, TCRneg/low, which bear a pro-inflammatory activation profile. Strikingly, we also found elevated levels of these undifferentiated T lymphocytes in the peripheral blood of patients in severe cardiac forms of chronic Chagas disease. Importantly, we provided evidence that migration of CD4−CD8− T cells from infected mouse thymus is due to sphingosine-1-phosphate receptor-1-dependent chemotaxis. These findings point to an important role for bioactive signaling sphingolipids in the thymic escape of immature thymocytes to the periphery in Chagas disease.
Collapse
Affiliation(s)
- Ailin Lepletier
- Laboratory on Thymus Research, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Liliane de Almeida
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Santos
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luzia da Silva Sampaio
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bruno Paredes
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Juan Beloscar
- Servicio de Clínica Médica, Hospital J.B. Iturraspe, Santa Fe, Argentina
| | - Oscar Bottasso
- Servicio de Clínica Médica, Hospital J.B. Iturraspe, Santa Fe, Argentina
| | - Marcelo Einicker-Lamas
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Rosa Pérez
- Institute of Immunology, National University of Rosario, Rosario, Argentina
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Alexandre Morrot
- Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
27
|
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
|
28
|
Lepletier A, de Carvalho VF, e Silva PMR, Villar S, Pérez AR, Savino W, Morrot A. Trypanosoma cruzi disrupts thymic homeostasis by altering intrathymic and systemic stress-related endocrine circuitries. PLoS Negl Trop Dis 2013; 7:e2470. [PMID: 24324845 PMCID: PMC3852165 DOI: 10.1371/journal.pntd.0002470] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/27/2013] [Indexed: 01/01/2023] Open
Abstract
We have previously shown that experimental infection caused by Trypanosoma cruzi
is associated with changes in the hypothalamus-pituitary-adrenal axis. Increased glucocorticoid (GC)
levels are believed to be protective against the effects of acute stress during infection but result
in depletion of CD4+CD8+ thymocytes by apoptosis, driving to thymic
atrophy. However, very few data are available concerning prolactin (PRL), another stress-related
hormone, which seems to be decreased during T. cruzi infection. Considering the
immunomodulatory role of PRL upon the effects caused by GC, we investigated if intrathymic
cross-talk between GC and PRL receptors (GR and PRLR, respectively) might influence T.
cruzi-induced thymic atrophy. Using an acute experimental model, we observed changes in
GR/PRLR cross-activation related with the survival of CD4+CD8+
thymocytes during infection. These alterations were closely related with systemic changes,
characterized by a stress hormone imbalance, with progressive GC augmentation simultaneously to PRL
reduction. The intrathymic hormone circuitry exhibited an inverse modulation that seemed to
counteract the GC-related systemic deleterious effects. During infection, adrenalectomy protected
the thymus from the increase in apoptosis ratio without changing PRL levels, whereas an additional
inhibition of circulating PRL accelerated the thymic atrophy and led to an increase in
corticosterone systemic levels. These results demonstrate that the PRL impairment during infection
is not caused by the increase of corticosterone levels, but the opposite seems to occur.
Accordingly, metoclopramide (MET)-induced enhancement of PRL secretion protected thymic atrophy in
acutely infected animals as well as the abnormal export of immature and potentially autoreactive
CD4+CD8+ thymocytes to the periphery. In conclusion, our findings
clearly show that Trypanosoma cruzi subverts mouse thymus homeostasis by altering
intrathymic and systemic stress-related endocrine circuitries with major consequences upon the
normal process of intrathymic T cell development. It is currently estimated that 90 million people in America are exposed to T.
cruzi infection, the causative agent of Chagas disease. Despite the mortality and
morbidity, this infection is yet considered a neglected disease, due to the lack of effective, safe,
and affordable pharmaceuticals for controlling it. T. cruzi leads to
immunosuppression of the T cell compartment and to chronic cardiac inflammation, which seems to be
associated with the disruption of thymic homeostasis. Thymus atrophy, characteristic of acute
infection, is mainly associated with the loss of immature CD4+CD8+
thymocytes, which in turn is associated with increased corticosterone systemic levels, together with
their premature export to the periphery as potential autorreactive cells. Although being deleterious
to the thymus, GCs are protective during this infection, for avoiding an exacerbated
pro-inflammatory response. Here we demonstrate that the increase of GCs in plasma is related to the
impairment of PRL systemic levels. The intrathymic hormonal circuitry is also altered during
infection and an imbalance of the cross-talk involving GR and PRL is related with
CD4+CD8+ depletion. The partial restoration of PRL levels prevented
thymus atrophy of infected mice, thus partially reverting the T. cruzi-induced
subversion of the organ, ultimately reestablishing thymus homeostasis.
Collapse
Affiliation(s)
- Ailin Lepletier
- Laboratory of Thymus Research, Oswaldo Cruz Institute,
Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | | | - Silvina Villar
- Institute of Immunology, Faculty of Medical Sciences,
National University of Rosario and CONICET, Rosario, Argentina
| | - Ana Rosa Pérez
- Institute of Immunology, Faculty of Medical Sciences,
National University of Rosario and CONICET, Rosario, Argentina
| | - Wilson Savino
- Laboratory of Thymus Research, Oswaldo Cruz Institute,
Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- * E-mail: ,
| | - Alexandre Morrot
- Laboratory of Thymus Research, Oswaldo Cruz Institute,
Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Immunobiology, Paulo de Goes Institute of
Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
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
|
30
|
Perniola R. Expression of the autoimmune regulator gene and its relevance to the mechanisms of central and peripheral tolerance. Clin Dev Immunol 2012; 2012:207403. [PMID: 23125865 PMCID: PMC3485510 DOI: 10.1155/2012/207403] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 08/26/2012] [Accepted: 09/11/2012] [Indexed: 01/12/2023]
Abstract
The autoimmune polyendocrine syndrome type 1 (APS-1) is a monogenic disease due to pathogenic variants occurring in the autoimmune regulator (AIRE) gene. Its related protein, AIRE, activates the transcription of genes encoding for tissue-specific antigens (TsAgs) in a subset of medullary thymic epithelial cells: the presentation of TsAgs to the maturating thymocytes induces the apoptosis of the autoreactive clones and constitutes the main form of central tolerance. Dysregulation of thymic AIRE expression in genetically transmitted and acquired diseases other than APS-1 may contribute to further forms of autoimmunity. As AIRE and its murine homolog are also expressed in the secondary lymphoid organs, the extent and relevance of AIRE participation in the mechanisms of peripheral tolerance need to be thoroughly defined.
Collapse
Affiliation(s)
- Roberto Perniola
- Neonatal Intensive Care, Department of Pediatrics, V. Fazzi Regional Hospital, Piazza F. Muratore, 73100 Lecce, Italy.
| |
Collapse
|
31
|
Borges M, Barreira-Silva P, Flórido M, Jordan MB, Correia-Neves M, Appelberg R. Molecular and cellular mechanisms of Mycobacterium avium-induced thymic atrophy. THE JOURNAL OF IMMUNOLOGY 2012; 189:3600-8. [PMID: 22922815 DOI: 10.4049/jimmunol.1201525] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Thymic atrophy has been described as a consequence of infection by several pathogens and shown to be induced through diverse mechanisms. Using the mouse model of Mycobacterium avium infection, we show in this study that the production of NO from IFN-γ-activated macrophages plays a major role in mycobacterial infection-induced thymic atrophy. Our results show that disseminated infection with a highly virulent strain of M. avium, but not with a low-virulence strain, led to a progressive thymic atrophy. Thymic involution was prevented in genetically manipulated mice unable to produce IFN-γ or the inducible NO synthase. In addition, mice with a selective impairment of IFN-γ signaling in macrophages were similarly protected from infection-induced thymic atrophy. A slight increase in the concentration of corticosterone was found in mice infected with the highly virulent strain, and thymocytes presented an increased susceptibility to dexamethasone-induced death during disseminated infection. The administration of an antagonist of glucocorticoid receptors partially reverted the infection-induced thymic atrophy. We observed a reduction in all thymocyte populations analyzed, including the earliest thymic precursors, suggesting a defect during thymic colonization by T cell precursors and/or during the differentiation of these cells in the bone marrow in addition to local demise of thymic cells. Our data suggest a complex picture underlying thymic atrophy during infection by M. avium with the participation of locally produced NO, endogenous corticosteroid activity, and reduced bone marrow seeding.
Collapse
Affiliation(s)
- Margarida Borges
- Institute for Molecular and Cell Biology, University of Porto, 4150-180 Porto, Portugal
| | | | | | | | | | | |
Collapse
|
32
|
Pérez AR, Morrot A, Berbert LR, Terra-Granado E, Savino W. Extrathymic CD4+CD8+ lymphocytes in Chagas disease: possible relationship with an immunoendocrine imbalance. Ann N Y Acad Sci 2012; 1262:27-36. [DOI: 10.1111/j.1749-6632.2012.06627.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
33
|
Pérez AR, Berbert LR, Lepletier A, Revelli S, Bottasso O, Silva-Barbosa SD, Savino W. TNF-α is involved in the abnormal thymocyte migration during experimental Trypanosoma cruzi infection and favors the export of immature cells. PLoS One 2012; 7:e34360. [PMID: 22461911 PMCID: PMC3312912 DOI: 10.1371/journal.pone.0034360] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 02/27/2012] [Indexed: 11/28/2022] Open
Abstract
Previous studies revealed a significant production of inflammatory cytokines together with severe thymic atrophy and thymocyte migratory disturbances during experimental Chagas disease. Migratory activity of thymocytes and mature T cells seem to be finely tuned by cytokines, chemokines and extracellular matrix (ECM) components. Systemic TNF-α is enhanced during infection and appears to be crucial in the response against the parasite. However, it also seems to be involved in disease pathology, since it is implicated in the arrival of T cells to effector sites, including the myocardium. Herein, we analyzed the role of TNF-α in the migratory activity of thymocytes in Trypanosoma cruzi (T. cruzi) acutely-infected mice. We found increased expression and deposition of TNF-α in the thymus of infected animals compared to controls, accompanied by increased co-localization of fibronectin, a cell migration-related ECM molecule, whose contents in the thymus of infected mice is also augmented. In-vivo studies showed an enhanced export of thymocytes in T. cruzi-infected mice, as ascertained by intrathymic injection of FITC alone or in combination with TNF-α. The increase of immature CD4+CD8+ T cells in secondary lymphoid organs was even more clear-cut when TNF-α was co-injected with FITC. Ex-vivo transmigration assays also revealed higher number of migrating cells when TNF-α was added onto fibronectin lattices, with higher input of all thymocyte subsets, including immature CD4+CD8+. Infected animals also exhibit enhanced levels of expression of both mRNA TNF-α receptors in the CD4+CD8+ subpopulation. Our findings suggest that in T. cruzi acute infection, when TNF-α is complexed with fibronectin, it favours the altered migration of thymocytes, promoting the release of mature and immature T cells to different compartments of the immune system. Conceptually, this work reinforces the notion that thymocyte migration is a multivectorial biological event in health and disease, and that TNF-α is a further player in the process.
Collapse
Affiliation(s)
- Ana Rosa Pérez
- Faculty of Medical Sciences, Institute of Immunology, National University of Rosario, Rosario, Argentina.
| | | | | | | | | | | | | |
Collapse
|
34
|
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
|
35
|
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
|